Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
Nature ; 485(7399): 512-6, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22622580

RESUMEN

Human apolipoprotein E has three isoforms: APOE2, APOE3 and APOE4. APOE4 is a major genetic risk factor for Alzheimer's disease and is associated with Down's syndrome dementia and poor neurological outcome after traumatic brain injury and haemorrhage. Neurovascular dysfunction is present in normal APOE4 carriers and individuals with APOE4-associated disorders. In mice, lack of Apoe leads to blood-brain barrier (BBB) breakdown, whereas APOE4 increases BBB susceptibility to injury. How APOE genotype affects brain microcirculation remains elusive. Using different APOE transgenic mice, including mice with ablation and/or inhibition of cyclophilin A (CypA), here we show that expression of APOE4 and lack of murine Apoe, but not APOE2 and APOE3, leads to BBB breakdown by activating a proinflammatory CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes. This, in turn, leads to neuronal uptake of multiple blood-derived neurotoxic proteins, and microvascular and cerebral blood flow reductions. We show that the vascular defects in Apoe-deficient and APOE4-expressing mice precede neuronal dysfunction and can initiate neurodegenerative changes. Astrocyte-secreted APOE3, but not APOE4, suppressed the CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes through a lipoprotein receptor. Our data suggest that CypA is a key target for treating APOE4-mediated neurovascular injury and the resulting neuronal dysfunction and degeneration.


Asunto(s)
Apolipoproteínas E/metabolismo , Barrera Hematoencefálica/fisiología , Circulación Cerebrovascular/fisiología , Ciclofilina A/metabolismo , Animales , Apolipoproteína E2/deficiencia , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E3/deficiencia , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/deficiencia , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Ciclofilina A/antagonistas & inhibidores , Ciclofilina A/deficiencia , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Microcirculación , FN-kappa B/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Pericitos/metabolismo
3.
J Neurosci ; 35(31): 11034-44, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26245965

RESUMEN

The glymphatic pathway expedites clearance of waste, including soluble amyloid ß (Aß) from the brain. Transport through this pathway is controlled by the brain's arousal level because, during sleep or anesthesia, the brain's interstitial space volume expands (compared with wakefulness), resulting in faster waste removal. Humans, as well as animals, exhibit different body postures during sleep, which may also affect waste removal. Therefore, not only the level of consciousness, but also body posture, might affect CSF-interstitial fluid (ISF) exchange efficiency. We used dynamic-contrast-enhanced MRI and kinetic modeling to quantify CSF-ISF exchange rates in anesthetized rodents' brains in supine, prone, or lateral positions. To validate the MRI data and to assess specifically the influence of body posture on clearance of Aß, we used fluorescence microscopy and radioactive tracers, respectively. The analysis showed that glymphatic transport was most efficient in the lateral position compared with the supine or prone positions. In the prone position, in which the rat's head was in the most upright position (mimicking posture during the awake state), transport was characterized by "retention" of the tracer, slower clearance, and more CSF efflux along larger caliber cervical vessels. The optical imaging and radiotracer studies confirmed that glymphatic transport and Aß clearance were superior in the lateral and supine positions. We propose that the most popular sleep posture (lateral) has evolved to optimize waste removal during sleep and that posture must be considered in diagnostic imaging procedures developed in the future to assess CSF-ISF transport in humans. SIGNIFICANCE STATEMENT: The rodent brain removes waste better during sleep or anesthesia compared with the awake state. Animals exhibit different body posture during the awake and sleep states, which might affect the brain's waste removal efficiency. We investigated the influence of body posture on brainwide transport of inert tracers of anesthetized rodents. The major finding of our study was that waste, including Aß, removal was most efficient in the lateral position (compared with the prone position), which mimics the natural resting/sleeping position of rodents. Although our finding awaits testing in humans, we speculate that the lateral position during sleep has advantage with regard to the removal of waste products including Aß, because clinical studies have shown that sleep drives Aß clearance from the brain.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Postura/fisiología , Animales , Transporte Biológico , Femenino , Imagen por Resonancia Magnética , Ratones , Ratas , Ratas Sprague-Dawley
4.
J Neurosci ; 35(2): 518-26, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589747

RESUMEN

The nonspecific and variable presentation of traumatic brain injury (TBI) has motivated an intense search for blood-based biomarkers that can objectively predict the severity of injury. However, it is not known how cytosolic proteins released from traumatized brain tissue reach the peripheral blood. Here we show in a murine TBI model that CSF movement through the recently characterized glymphatic pathway transports biomarkers to blood via the cervical lymphatics. Clinically relevant manipulation of glymphatic activity, including sleep deprivation and cisternotomy, suppressed or eliminated TBI-induced increases in serum S100ß, GFAP, and neuron specific enolase. We conclude that routine TBI patient management may limit the clinical utility of blood-based biomarkers because their brain-to-blood transport depends on glymphatic activity.


Asunto(s)
Lesiones Encefálicas/metabolismo , Líquido Extracelular/metabolismo , Tasa de Depuración Metabólica , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/sangre , Lesiones Encefálicas/líquido cefalorraquídeo , Femenino , Proteína Ácida Fibrilar de la Glía/sangre , Proteína Ácida Fibrilar de la Glía/líquido cefalorraquídeo , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/sangre , Subunidad beta de la Proteína de Unión al Calcio S100/líquido cefalorraquídeo , Privación de Sueño/sangre , Privación de Sueño/líquido cefalorraquídeo , Privación de Sueño/metabolismo
5.
Neurobiol Dis ; 93: 215-25, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27234656

RESUMEN

Glymphatic transport, defined as cerebrospinal fluid (CSF) peri-arterial inflow into brain, and interstitial fluid (ISF) clearance, is reduced in the aging brain. However, it is unclear whether glymphatic transport affects the distribution of soluble Aß in Alzheimer's disease (AD). In wild type mice, we show that Aß40 (fluorescently labeled Aß40 or unlabeled Aß40), was distributed from CSF to brain, via the peri-arterial space, and associated with neurons. In contrast, Aß42 was mostly restricted to the peri-arterial space due mainly to its greater propensity to oligomerize when compared to Aß40. Interestingly, pretreatment with Aß40 in the CSF, but not Aß42, reduced CSF transport into brain. In APP/PS1 mice, a model of AD, with and without extensive amyloid-ß deposits, glymphatic transport was reduced, due to the accumulation of toxic Aß species, such as soluble oligomers. CSF-derived Aß40 co-localizes with existing endogenous vascular and parenchymal amyloid-ß plaques, and thus, may contribute to the progression of both cerebral amyloid angiopathy and parenchymal Aß accumulation. Importantly, glymphatic failure preceded significant amyloid-ß deposits, and thus, may be an early biomarker of AD. By extension, restoring glymphatic inflow and ISF clearance are potential therapeutic targets to slow the onset and progression of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Placa Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Transporte Biológico/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones Endogámicos C57BL
6.
Proc Natl Acad Sci U S A ; 110(36): 14771-6, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959870

RESUMEN

Whereas amyloid-ß (Aß) accumulates in the brain of normal animals dosed with low levels of copper (Cu), the mechanism is not completely known. Cu could contribute to Aß accumulation by altering its clearance and/or its production. Because Cu homeostasis is altered in transgenic mice overexpressing Aß precursor protein (APP), the objective of this study was to elucidate the mechanism of Cu-induced Aß accumulation in brains of normal mice and then to explore Cu's effects in a mouse model of Alzheimer's disease. In aging mice, accumulation of Cu in brain capillaries was associated with its reduction in low-density lipoprotein receptor-related protein 1 (LRP1), an Aß transporter, and higher brain Aß levels. These effects were reproduced by chronic dosing with low levels of Cu via drinking water without changes in Aß synthesis or degradation. In human brain endothelial cells, Cu, at its normal labile levels, caused LRP1-specific down-regulation by inducing its nitrotyrosination and subsequent proteosomal-dependent degradation due in part to Cu/cellular prion protein/LRP1 interaction. In APP(sw/0) mice, Cu not only down-regulated LRP1 in brain capillaries but also increased Aß production and neuroinflammation because Cu accumulated in brain capillaries and, unlike in control mice, in the parenchyma. Thus, we have demonstrated that Cu's effect on brain Aß homeostasis depends on whether it is accumulated in the capillaries or in the parenchyma. These findings should provide unique insights into preventative and/or therapeutic approaches to control neurotoxic Aß levels in the aging brain.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/efectos de los fármacos , Cobre/farmacología , Homeostasis/efectos de los fármacos , Factores de Edad , Péptidos beta-Amiloides/farmacocinética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Western Blotting , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Capilares/efectos de los fármacos , Capilares/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cobre/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Radioisótopos de Yodo/farmacocinética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Factores de Tiempo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
7.
J Neurosci ; 34(49): 16180-93, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471560

RESUMEN

Traumatic brain injury (TBI) is an established risk factor for the early development of dementia, including Alzheimer's disease, and the post-traumatic brain frequently exhibits neurofibrillary tangles comprised of aggregates of the protein tau. We have recently defined a brain-wide network of paravascular channels, termed the "glymphatic" pathway, along which CSF moves into and through the brain parenchyma, facilitating the clearance of interstitial solutes, including amyloid-ß, from the brain. Here we demonstrate in mice that extracellular tau is cleared from the brain along these paravascular pathways. After TBI, glymphatic pathway function was reduced by ∼60%, with this impairment persisting for at least 1 month post injury. Genetic knock-out of the gene encoding the astroglial water channel aquaporin-4, which is importantly involved in paravascular interstitial solute clearance, exacerbated glymphatic pathway dysfunction after TBI and promoted the development of neurofibrillary pathology and neurodegeneration in the post-traumatic brain. These findings suggest that chronic impairment of glymphatic pathway function after TBI may be a key factor that renders the post-traumatic brain vulnerable to tau aggregation and the onset of neurodegeneration.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Líquido Cefalorraquídeo/metabolismo , Líquido Extracelular/metabolismo , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Proteínas tau/metabolismo , Animales , Acuaporina 4/genética , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/genética
8.
Ann Neurol ; 76(6): 845-61, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25204284

RESUMEN

OBJECTIVE: In the brain, protein waste removal is partly performed by paravascular pathways that facilitate convective exchange of water and soluble contents between cerebrospinal fluid (CSF) and interstitial fluid (ISF). Several lines of evidence suggest that bulk flow drainage via the glymphatic system is driven by cerebrovascular pulsation, and is dependent on astroglial water channels that line paravascular CSF pathways. The objective of this study was to evaluate whether the efficiency of CSF-ISF exchange and interstitial solute clearance is impaired in the aging brain. METHODS: CSF-ISF exchange was evaluated by in vivo and ex vivo fluorescence microscopy and interstitial solute clearance was evaluated by radiotracer clearance assays in young (2-3 months), middle-aged (10-12 months), and old (18-20 months) wild-type mice. The relationship between age-related changes in the expression of the astrocytic water channel aquaporin-4 (AQP4) and changes in glymphatic pathway function was evaluated by immunofluorescence. RESULTS: Advancing age was associated with a dramatic decline in the efficiency of exchange between the subarachnoid CSF and the brain parenchyma. Relative to the young, clearance of intraparenchymally injected amyloid-ß was impaired by 40% in the old mice. A 27% reduction in the vessel wall pulsatility of intracortical arterioles and widespread loss of perivascular AQP4 polarization along the penetrating arteries accompanied the decline in CSF-ISF exchange. INTERPRETATION: We propose that impaired glymphatic clearance contributes to cognitive decline among the elderly and may represent a novel therapeutic target for the treatment of neurodegenerative diseases associated with accumulation of misfolded protein aggregates.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Circulación Cerebrovascular/fisiología , Tasa de Depuración Metabólica/fisiología , Envejecimiento/patología , Animales , Acuaporina 4/metabolismo , Encéfalo/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neuroglía/metabolismo , Neuroglía/patología
9.
Proc Natl Acad Sci U S A ; 109(38): 15502-7, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22927427

RESUMEN

The apolipoprotein E (APOE)-ε4 allele is the strongest genetic risk factor for late-onset, sporadic Alzheimer's disease, likely increasing risk by altering amyloid-ß (Aß) accumulation. We recently demonstrated that the low-density lipoprotein receptor (LDLR) is a major apoE receptor in the brain that strongly regulates amyloid plaque deposition. In the current study, we sought to understand the mechanism by which LDLR regulates Aß accumulation by altering Aß clearance from brain interstitial fluid. We hypothesized that increasing LDLR levels enhances blood-brain barrier-mediated Aß clearance, thus leading to reduced Aß accumulation. Using the brain Aß efflux index method, we found that blood-brain barrier-mediated clearance of exogenously administered Aß is enhanced with LDLR overexpression. We next developed a method to directly assess the elimination of centrally derived, endogenous Aß into the plasma of mice using an anti-Aß antibody that prevents degradation of plasma Aß, allowing its rate of appearance from the brain to be measured. Using this plasma Aß accumulation technique, we found that LDLR overexpression enhances brain-to-blood Aß transport. Together, our results suggest a unique mechanism by which LDLR regulates brain-to-blood Aß clearance, which may serve as a useful therapeutic avenue in targeting Aß clearance from the brain.


Asunto(s)
Amiloidosis/metabolismo , Apolipoproteína E4/genética , Receptores de LDL/biosíntesis , Alelos , Péptidos beta-Amiloides/metabolismo , Animales , Barrera Hematoencefálica , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Insulina/metabolismo , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Microdiálisis , Transgenes
10.
J Neurosci ; 33(46): 18190-9, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24227727

RESUMEN

CSF from the subarachnoid space moves rapidly into the brain along paravascular routes surrounding penetrating cerebral arteries, exchanging with brain interstitial fluid (ISF) and facilitating the clearance of interstitial solutes, such as amyloid ß, in a pathway that we have termed the "glymphatic" system. Prior reports have suggested that paravascular bulk flow of CSF or ISF may be driven by arterial pulsation. However, cerebral arterial pulsation could not be directly assessed. In the present study, we use in vivo two-photon microscopy in mice to visualize vascular wall pulsatility in penetrating intracortical arteries. We observed that unilateral ligation of the internal carotid artery significantly reduced arterial pulsatility by ~50%, while systemic administration of the adrenergic agonist dobutamine increased pulsatility of penetrating arteries by ~60%. When paravascular CSF-ISF exchange was evaluated in real time using in vivo two-photon and ex vivo fluorescence imaging, we observed that internal carotid artery ligation slowed the rate of paravascular CSF-ISF exchange, while dobutamine increased the rate of paravascular CSF-ISF exchange. These findings demonstrate that cerebral arterial pulsatility is a key driver of paravascular CSF influx into and through the brain parenchyma, and suggest that changes in arterial pulsatility may contribute to accumulation and deposition of toxic solutes, including amyloid ß, in the aging brain.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Encéfalo/irrigación sanguínea , Encéfalo/fisiología , Arterias Cerebrales/fisiología , Líquido Cefalorraquídeo/fisiología , Animales , Química Encefálica/fisiología , Arterias Cerebrales/química , Líquido Cefalorraquídeo/química , Líquido Extracelular/química , Líquido Extracelular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía de Fluorescencia por Excitación Multifotónica/métodos
11.
Nat Med ; 13(9): 1029-31, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17694066

RESUMEN

Low-density lipoprotein receptor-related protein-1 (LRP) on brain capillaries clears amyloid beta-peptide (Abeta) from brain. Here, we show that soluble circulating LRP (sLRP) provides key endogenous peripheral 'sink' activity for Abeta in humans. Recombinant LRP cluster IV (LRP-IV) bound Abeta in plasma in mice and Alzheimer's disease-affected humans with compromised sLRP-mediated Abeta binding, and reduced Abeta-related pathology and dysfunction in a mouse model of Alzheimer disease, suggesting that LRP-IV can effectively replace native sLRP and clear Abeta.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Capilares/fisiología , Circulación Cerebrovascular/fisiología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Heterocigoto , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/sangre , Ratones
12.
J Transl Med ; 11: 107, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23635358

RESUMEN

BACKGROUND: Neurodegenerative diseases such as Alzheimer's are associated with the aggregation of endogenous peptides and proteins that contribute to neuronal dysfunction and loss. The glymphatic system, a brain-wide perivascular pathway along which cerebrospinal fluid (CSF) and interstitial fluid (ISF) rapidly exchange, has recently been identified as a key contributor to the clearance of interstitial solutes from the brain, including amyloid ß. These findings suggest that measuring changes in glymphatic pathway function may be an important prognostic for evaluating neurodegenerative disease susceptibility or progression. However, no clinically acceptable approach to evaluate glymphatic pathway function in humans has yet been developed. METHODS: Time-sequenced ex vivo fluorescence imaging of coronal rat and mouse brain slices was performed at 30-180 min following intrathecal infusion of CSF tracer (Texas Red- dextran-3, MW 3 kD; FITC- dextran-500, MW 500 kD) into the cisterna magna or lumbar spine. Tracer influx into different brain regions (cortex, white matter, subcortical structures, and hippocampus) in rat was quantified to map the movement of CSF tracer following infusion along both routes, and to determine whether glymphatic pathway function could be evaluated after lumbar intrathecal infusion. RESULTS: Following lumbar intrathecal infusions, small molecular weight TR-d3 entered the brain along perivascular pathways and exchanged broadly with the brain ISF, consistent with the initial characterization of the glymphatic pathway in mice. Large molecular weight FITC-d500 remained confined to the perivascular spaces. Lumbar intrathecal infusions exhibited a reduced and delayed peak parenchymal fluorescence intensity compared to intracisternal infusions. CONCLUSION: Lumbar intrathecal contrast delivery is a clinically useful approach that could be used in conjunction with dynamic contrast enhanced MRI nuclear imaging to assess glymphatic pathway function in humans.


Asunto(s)
Encéfalo/irrigación sanguínea , Líquido Cefalorraquídeo/metabolismo , Inyecciones Espinales , Sondas Moleculares , Animales , Líquido Extracelular/metabolismo , Femenino , Presión Intracraneal , Vértebras Lumbares/metabolismo , Masculino , Ratones Endogámicos C57BL , Peso Molecular , Ratas Sprague-Dawley
13.
Nat Med ; 12(11): 1278-85, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17072311

RESUMEN

Brain hemorrhage is a serious complication of tissue plasminogen activator (tPA) therapy for ischemic stroke. Here we report that activated protein C (APC), a plasma serine protease with systemic anticoagulant, anti-inflammatory and antiapoptotic activities, and direct vasculoprotective and neuroprotective activities, blocks tPA-mediated brain hemorrhage after transient brain ischemia and embolic stroke in rodents. We show that APC inhibits a pro-hemorrhagic tPA-induced, NF-kappaB-dependent matrix metalloproteinase-9 pathway in ischemic brain endothelium in vivo and in vitro by acting through protease-activated receptor 1. The present findings suggest that APC may improve thrombolytic therapy for stroke, in part, by reducing tPA-mediated hemorrhage.


Asunto(s)
Hemorragias Intracraneales/prevención & control , Proteína C/farmacología , Activador de Tejido Plasminógeno/farmacología , Animales , Hemorragias Intracraneales/inducido químicamente , Hemorragias Intracraneales/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes/farmacología
14.
Front Neurosci ; 17: 1117845, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875642

RESUMEN

Introduction: There is clinical evidence of neurological manifestations in coronavirus disease-19 (COVID-19). However, it is unclear whether differences in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/spike protein (SP) uptake by cells of the cerebrovasculature contribute to significant viral uptake to cause these symptoms. Methods: Since the initial step in viral invasion is binding/uptake, we used fluorescently labeled wild type and mutant SARS-CoV-2/SP to study this process. Three cerebrovascular cell types were used (endothelial cells, pericytes, and vascular smooth muscle cells), in vitro. Results: There was differential SARS-CoV-2/SP uptake by these cell types. Endothelial cells had the least uptake, which may limit SARS-CoV-2 uptake into brain from blood. Uptake was time and concentration dependent, and mediated by angiotensin converting enzyme 2 receptor (ACE2), and ganglioside (mono-sialotetrahexasylganglioside, GM1) that is predominantly expressed in the central nervous system and the cerebrovasculature. SARS-CoV-2/SPs with mutation sites, N501Y, E484K, and D614G, as seen in variants of interest, were also differentially taken up by these cell types. There was greater uptake compared to that of the wild type SARS-CoV-2/SP, but neutralization with anti-ACE2 or anti-GM1 antibodies was less effective. Conclusion: The data suggested that in addition to ACE2, gangliosides are also an important entry point of SARS-CoV-2/SP into these cells. Since SARS-CoV-2/SP binding/uptake is the initial step in the viral penetration into cells, a longer exposure and higher titer are required for significant uptake into the normal brain. Gangliosides, including GM1, could be an additional potential SARS-CoV-2 and therapeutic target at the cerebrovasculature.

15.
bioRxiv ; 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37645819

RESUMEN

Tau protein blood levels dependent on its distribution to peripheral organs and possible elimination from the body. Thus, the peripheral distribution of CSF-derived tau protein was explored, especially since there is a transition to blood-based biomarkers and the emerging idea that tau pathology may spread beyond brain. Near infrared fluorescence (NIRF) was mainly used to analyze tau (tau-NIRF) distribution after its intracisternal or intravenous injection. There was a striking uptake of blood- or CSF-derived tau-NIRF protein by the skeletal structures, liver, small intestine (duodenum), gall bladder, kidneys, urinary bladder, lymph nodes, heart, and spleen. In aging and in older APP/PS1 mice, tau uptake in regions, such as the brain, liver, and skeleton, was increased. In bone (femur) injected tau protein was associated with integrin-binding sialoprotein (IBSP), a major non-collagenous glycoprotein that is associated with mineralization. Tau-NIRF was cleared slowly from CSF via mainly across the cribriform plate, and cervical lymph nodes. In brain, some of the CSF injected tau protein was associated with NeuN-positive and PDGFRý-positive cells, which may explain its retention. The presence of tau in the bladders suggested excretion routes of tau. CSF anti-tau antibody increased CSF tau clearance, while blood anti-tau antibody decreased tau accumulation in the femur but not in liver, kidney, and spleen. Thus, the data show a body-wide distribution and retention of CSF-derived tau protein, which increased with aging and in older APP/PS1 mice. Further work is needed to elucidate the relevance of tau accumulation in each organ to tauopathy.

16.
Blood ; 115(23): 4963-72, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20348395

RESUMEN

The anticoagulant factor protein S (PS) has direct cellular activities. Lack of PS in mice causes lethal coagulopathy, ischemic/thrombotic injuries, vascular dysgenesis, and blood-brain barrier (BBB) disruption with intracerebral hemorrhages. Thus, we hypothesized that PS maintains and/or enhances the BBB integrity. Using a BBB model with human brain endothelial cells, we show PS inhibits time- and dose-dependently (half maximal effective concentration [EC(50)] = 27 +/- 3 nM) oxygen/glucose deprivation-induced BBB breakdown, as demonstrated by measurements of the transmonolayer electrical resistance, permeability of endothelial monolayers to dextran (40 kDa), and rearrangement of F-actin toward the cortical cytoskeletal ring. Using Tyro-3, Axl, and Mer (TAM) receptor, tyrosine kinase silencing through RNA interference, specific N-terminus-blocking antibodies, Tyro3 phosphorylation, and Tyro3-, Axl- and Mer-deficient mouse brain endothelial cells, we show that Tyro3 mediates PS vasculoprotection. After Tyro3 ligation, PS activated sphingosine 1-phosphate receptor (S1P(1)), resulting in Rac1-dependent BBB protection. Using 2-photon in vivo imaging, we show that PS blocks postischemic BBB disruption in Tyro3(+/+), Axl(-/-), and Mer(-/-) mice, but not in Tyro3(-/-) mice or Tyro3(+/+) mice receiving low-dose W146, a S1P(1)-specific antagonist. Our findings indicate that PS protects the BBB integrity via Tyro3 and S1P(1), suggesting potentially novel treatments for neurovascular dysfunction resulting from hypoxic/ischemic BBB damage.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Proteína S/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Animales , Isquemia Encefálica/genética , Humanos , Ratones , Ratones Mutantes , Modelos Biológicos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fosforilación/genética , Proteína S/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato , Adulto Joven , Tirosina Quinasa c-Mer , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Tirosina Quinasa del Receptor Axl
17.
Nat Med ; 11(9): 959-65, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116430

RESUMEN

Neurovascular dysfunction substantially contributes to Alzheimer disease. Here, we show that transcriptional profiling of human brain endothelial cells (BECs) defines a subset of genes whose expression is age-independent but is considerably altered in Alzheimer disease, including the homeobox gene MEOX2 (also known as GAX), a regulator of vascular differentiation, whose expression is low in Alzheimer disease. By using viral-mediated MEOX2 gene silencing and transfer, we show that restoring expression of the protein it encodes, GAX, in BECs from individuals with Alzheimer disease stimulates angiogenesis, transcriptionally suppresses AFX1 forkhead transcription factor-mediated apoptosis and increases the levels of a major amyloid-beta peptide (Abeta) clearance receptor, the low-density lipoprotein receptor-related protein 1 (LRP), at the blood-brain barrier. In mice, deletion of Meox2 (also known as Gax) results in reductions in brain capillary density and resting cerebral blood flow, loss of the angiogenic response to hypoxia in the brain and an impaired Abeta efflux from brain caused by reduced LRP levels. The link of MEOX2 to neurovascular dysfunction in Alzheimer disease provides new mechanistic and therapeutic insights into this illness.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/fisiología , Genes Homeobox , Enfermedad de Alzheimer/metabolismo , Animales , Apoptosis , Células Cultivadas , Lóbulo Frontal/irrigación sanguínea , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Neovascularización Fisiológica/genética
18.
Nat Neurosci ; 11(4): 420-2, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18344992

RESUMEN

We report here that amyotrophic lateral sclerosis-linked superoxide dismutase 1 (SOD1) mutants with different biochemical characteristics disrupted the blood-spinal cord barrier in mice by reducing the levels of the tight junction proteins ZO-1, occludin and claudin-5 between endothelial cells. This resulted in microhemorrhages with release of neurotoxic hemoglobin-derived products, reductions in microcirculation and hypoperfusion. SOD1 mutant-mediated endothelial damage accumulated before motor neuron degeneration and the neurovascular inflammatory response occurred, indicating that it was a central contributor to disease initiation.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Barrera Hematoencefálica/metabolismo , Capilares/enzimología , Endotelio Vascular/metabolismo , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Barrera Hematoencefálica/patología , Capilares/patología , Claudina-5 , Progresión de la Enfermedad , Endotelio Vascular/patología , Hemorragia/patología , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Neuronas Motoras/patología , Degeneración Nerviosa/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Médula Espinal/irrigación sanguínea , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Uniones Estrechas/enzimología , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1
19.
J Clin Invest ; 118(12): 4002-13, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19033669

RESUMEN

Neurotoxic amyloid beta peptide (Abeta) accumulates in the brains of individuals with Alzheimer disease (AD). The APOE4 allele is a major risk factor for sporadic AD and has been associated with increased brain parenchymal and vascular amyloid burden. How apoE isoforms influence Abeta accumulation in the brain has, however, remained unclear. Here, we have shown that apoE disrupts Abeta clearance across the mouse blood-brain barrier (BBB) in an isoform-specific manner (specifically, apoE4 had a greater disruptive effect than either apoE3 or apoE2). Abeta binding to apoE4 redirected the rapid clearance of free Abeta40/42 from the LDL receptor-related protein 1 (LRP1) to the VLDL receptor (VLDLR), which internalized apoE4 and Abeta-apoE4 complexes at the BBB more slowly than LRP1. In contrast, apoE2 and apoE3 as well as Abeta-apoE2 and Abeta-apoE3 complexes were cleared at the BBB via both VLDLR and LRP1 at a substantially faster rate than Abeta-apoE4 complexes. Astrocyte-secreted lipo-apoE2, lipo-apoE3, and lipo-apoE4 as well as their complexes with Abeta were cleared at the BBB by mechanisms similar to those of their respective lipid-poor isoforms but at 2- to 3-fold slower rates. Thus, apoE isoforms differentially regulate Abeta clearance from the brain, and this might contribute to the effects of APOE genotype on the disease process in both individuals with AD and animal models of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Modelos Animales de Enfermedad , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Isoformas de Proteínas/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
20.
Nat Med ; 9(7): 907-13, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12808450

RESUMEN

Amyloid-beta peptide (Abeta) interacts with the vasculature to influence Abeta levels in the brain and cerebral blood flow, providing a means of amplifying the Abeta-induced cellular stress underlying neuronal dysfunction and dementia. Systemic Abeta infusion and studies in genetically manipulated mice show that Abeta interaction with receptor for advanced glycation end products (RAGE)-bearing cells in the vessel wall results in transport of Abeta across the blood-brain barrier (BBB) and expression of proinflammatory cytokines and endothelin-1 (ET-1), the latter mediating Abeta-induced vasoconstriction. Inhibition of RAGE-ligand interaction suppresses accumulation of Abeta in brain parenchyma in a mouse transgenic model. These findings suggest that vascular RAGE is a target for inhibiting pathogenic consequences of Abeta-vascular interactions, including development of cerebral amyloidosis.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Receptores Inmunológicos/metabolismo , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/farmacología , Animales , Encéfalo/irrigación sanguínea , Circulación Cerebrovascular , Citocinas/genética , Citocinas/metabolismo , Antagonistas de los Receptores de Endotelina , Endotelina-1/efectos de los fármacos , Endotelina-1/metabolismo , Humanos , Ratones , Ratones Transgénicos , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Transporte de Proteínas/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptor de Endotelina A
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA