Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 291
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 629(8013): 893-900, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632402

RESUMEN

The blood-brain barrier (BBB) protects the central nervous system from infections or harmful substances1; its impairment can lead to or exacerbate various diseases of the central nervous system2-4. However, the mechanisms of BBB disruption during infection and inflammatory conditions5,6 remain poorly defined. Here we find that activation of the pore-forming protein GSDMD by the cytosolic lipopolysaccharide (LPS) sensor caspase-11 (refs. 7-9), but not by TLR4-induced cytokines, mediates BBB breakdown in response to circulating LPS or during LPS-induced sepsis. Mice deficient in the LBP-CD14 LPS transfer and internalization pathway10-12 resist BBB disruption. Single-cell RNA-sequencing analysis reveals that brain endothelial cells (bECs), which express high levels of GSDMD, have a prominent response to circulating LPS. LPS acting on bECs primes Casp11 and Cd14 expression and induces GSDMD-mediated plasma membrane permeabilization and pyroptosis in vitro and in mice. Electron microscopy shows that this features ultrastructural changes in the disrupted BBB, including pyroptotic endothelia, abnormal appearance of tight junctions and vasculature detachment from the basement membrane. Comprehensive mouse genetic analyses, combined with a bEC-targeting adeno-associated virus system, establish that GSDMD activation in bECs underlies BBB disruption by LPS. Delivery of active GSDMD into bECs bypasses LPS stimulation and opens the BBB. In CASP4-humanized mice, Gram-negative Klebsiella pneumoniae infection disrupts the BBB; this is blocked by expression of a GSDMD-neutralizing nanobody in bECs. Our findings outline a mechanism for inflammatory BBB breakdown, and suggest potential therapies for diseases of the central nervous system associated with BBB impairment.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Células Endoteliales , Gasderminas , Inflamación , Animales , Femenino , Humanos , Masculino , Ratones , Membrana Basal/metabolismo , Membrana Basal/ultraestructura , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/ultraestructura , Barrera Hematoencefálica/virología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/ultraestructura , Caspasas Iniciadoras/metabolismo , Dependovirus , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Gasderminas/antagonistas & inhibidores , Gasderminas/metabolismo , Inflamación/patología , Inflamación/metabolismo , Klebsiella pneumoniae/fisiología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/sangre , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Piroptosis , Sepsis/metabolismo , Sepsis/patología , Sepsis/microbiología , Análisis de la Célula Individual , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura
2.
Proc Natl Acad Sci U S A ; 117(44): 27667-27675, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33087571

RESUMEN

Chronic neurodegeneration in survivors of traumatic brain injury (TBI) is a major cause of morbidity, with no effective therapies to mitigate this progressive and debilitating form of nerve cell death. Here, we report that pharmacologic restoration of the blood-brain barrier (BBB), 12 mo after murine TBI, is associated with arrested axonal neurodegeneration and cognitive recovery, benefits that persisted for months after treatment cessation. Recovery was achieved by 30 d of once-daily administration of P7C3-A20, a compound that stabilizes cellular energy levels. Four months after P7C3-A20, electron microscopy revealed full repair of TBI-induced breaks in cortical and hippocampal BBB endothelium. Immunohistochemical staining identified additional benefits of P7C3-A20, including restoration of normal BBB endothelium length, increased brain capillary pericyte density, increased expression of BBB tight junction proteins, reduced brain infiltration of immunoglobulin, and attenuated neuroinflammation. These changes were accompanied by cessation of TBI-induced chronic axonal degeneration. Specificity for P7C3-A20 action on the endothelium was confirmed by protection of cultured human brain microvascular endothelial cells from hydrogen peroxide-induced cell death, as well as preservation of BBB integrity in mice after exposure to toxic levels of lipopolysaccharide. P7C3-A20 also protected mice from BBB degradation after acute TBI. Collectively, our results provide insights into the pathophysiologic mechanisms behind chronic neurodegeneration after TBI, along with a putative treatment strategy. Because TBI increases the risks of other forms of neurodegeneration involving BBB deterioration (e.g., Alzheimer's disease, Parkinson's disease, vascular dementia, chronic traumatic encephalopathy), P7C3-A20 may have widespread clinical utility in the setting of neurodegenerative conditions.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Carbazoles/farmacología , Cognición/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/ultraestructura , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Carbazoles/uso terapéutico , Células Cultivadas , Enfermedad Crónica/tratamiento farmacológico , Cognición/fisiología , Modelos Animales de Enfermedad , Células Endoteliales , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Masculino , Ratones , Microscopía Electrónica , Microvasos/citología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Fármacos Neuroprotectores/uso terapéutico , Cultivo Primario de Células , Sobrevivientes
3.
Exp Cell Res ; 402(2): 112576, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33798592

RESUMEN

The brain vasculature has several specific features, one of them being the blood-brain barrier (BBB), which supports and protects the brain by allowing for the passage of oxygen and nutrients, while at the same time preventing passage of pathogens and toxins. The BBB also prevents efficient delivery of drugs to the brain, e.g. for treatment of brain tumors. In the murine brain, perivascular fibroblasts were recently identified as a novel potential constituent of the BBB. Here we present the existence of human cells that could be the equivalent to the murine brain perivascular fibroblasts. Using RNA sequencing, we show a similar transcriptomic profile of cultured human brain cells and murine perivascular fibroblasts. These data open up a window for new hypotheses on cell types involved in human CNS diseases.


Asunto(s)
Encéfalo/ultraestructura , Linaje de la Célula/genética , Sistema Nervioso Central/ultraestructura , Fibroblastos/metabolismo , Animales , Transporte Biológico/genética , Barrera Hematoencefálica/ultraestructura , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Ratones
4.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35054928

RESUMEN

The brain capillary endothelium is highly regulatory, maintaining the chemical stability of the brain's microenvironment. The role of cytoskeletal proteins in tethering nanotubules (TENTs) during barrier-genesis was investigated using the established immortalized mouse brain endothelial cell line (bEnd5) as an in vitro blood-brain barrier (BBB) model. The morphology of bEnd5 cells was evaluated using both high-resolution scanning electron microscopy and immunofluorescence to evaluate treatment with depolymerizing agents Cytochalasin D for F-actin filaments and Nocodazole for α-tubulin microtubules. The effects of the depolymerizing agents were investigated on bEnd5 monolayer permeability by measuring the transendothelial electrical resistance (TEER). The data endorsed that during barrier-genesis, F-actin and α-tubulin play a cytoarchitectural role in providing both cell shape dynamics and cytoskeletal structure to TENTs forming across the paracellular space to provide cell-cell engagement. Western blot analysis of the treatments suggested a reduced expression of both proteins, coinciding with a reduction in the rates of cellular proliferation and decreased TEER. The findings endorsed that TENTs provide alignment of the paracellular (PC) spaces and tight junction (TJ) zones to occlude bEnd5 PC spaces. The identification of specific cytoskeletal structures in TENTs endorsed the postulate of their indispensable role in barrier-genesis and the maintenance of regulatory permeability across the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Proteínas del Citoesqueleto/metabolismo , Actinas/metabolismo , Animales , Biomarcadores , Línea Celular , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Técnica del Anticuerpo Fluorescente , Expresión Génica , Ratones , Nocodazol/farmacología , Permeabilidad/efectos de los fármacos
5.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35054957

RESUMEN

Nanoparticles with oligonucleotides bound to the outside or incorporated into the matrix can be used for gene editing or to modulate gene expression in the CNS. These nanocarriers are usually optimised for transfection of neurons or glia. They can also facilitate transcytosis across the brain endothelium to circumvent the blood-brain barrier. This review examines the different formulations of nanocarriers and their oligonucleotide cargoes, in relation to their ability to enter the brain and modulate gene expression or disease. The size of the nanocarrier is critical in determining the rate of clearance from the plasma as well as the intracellular routes of endothelial transcytosis. The surface charge is important in determining how it interacts with the endothelium and the target cell. The structure of the oligonucleotide affects its stability and rate of degradation, while the chemical formulation of the nanocarrier primarily controls the location and rate of cargo release. Due to the major anatomical differences between humans and animal models of disease, successful gene therapy with oligonucleotides in humans has required intrathecal injection. In animal models, some progress has been made with intraventricular or intravenous injection of oligonucleotides on nanocarriers. However, getting significant amounts of nanocarriers across the blood-brain barrier in humans will likely require targeting endothelial solute carriers or vesicular transport systems.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Nanopartículas , Oligonucleótidos/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Central/metabolismo , Fenómenos Químicos , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Oro , Humanos , Nanopartículas del Metal , Tamaño de la Partícula
6.
Am J Physiol Heart Circ Physiol ; 320(2): H535-H548, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33275518

RESUMEN

Preeclampsia is characterized by increases in blood pressure and proteinuria in late pregnancy, and neurological symptoms can appear in the form of headaches, blurred vision, cerebral edema, and, in the most severe cases, seizures (eclampsia). The causes for these cerebral manifestations remain unknown, so the use of animal models that mimic preeclampsia is essential to understanding its pathogenesis. The Dahl salt-sensitive (Dahl SS/jr) rat model develops spontaneous preeclampsia superimposed on chronic hypertension; therefore, we hypothesized that the Dahl SS/jr rat would display cerebrovascular features similar to those seen in human preeclampsia. Furthermore, we predicted that this model would allow for the identification of mechanisms underlying these changes. The pregnant Dahl SS/jr rat displayed increased cerebral edema and blood-brain barrier disruption despite tighter control of cerebral blood flow autoregulation and vascular smooth muscle myogenic tone. Analysis of cerebral endothelial cell morphology revealed increased opening of tight junctions, basement membrane dissolution, and vesicle formation. RNAseq analysis identified that genes related to endothelial cell tight junctions and blood-brain barrier integrity were differentially expressed in cerebral vessels from pregnant Dahl SS/jr compared with healthy pregnant Sprague Dawley rats. Overall, our data reveal new insights into mechanisms involved in the cerebrovascular dysfunction of preeclampsia.NEW & NOTEWORTHY This study uses the Dahl SS/jr rat as a preclinical model of spontaneous superimposed preeclampsia to demonstrate uncoupling of cerebral vascular permeability and blood-brain barrier disruption from cerebral blood flow autoregulatory dysfunction and myogenic tone. Additionally, the data presented in this study lay the foundational framework on which future experiments assessing specific transcellular transport components such as individual transporter protein expression and components of the vesicular transport system (caveolae) can be built to help reveal a potential direct mechanistic insight into the causes of cerebrovascular complications during preeclamptic pregnancies.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Edema Encefálico/patología , Permeabilidad Capilar , Células Endoteliales/ultraestructura , Preeclampsia/patología , Animales , Membrana Basal/ultraestructura , Barrera Hematoencefálica/ultraestructura , Edema Encefálico/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Femenino , Preeclampsia/metabolismo , Embarazo , Ratas , Ratas Endogámicas Dahl , Ratas Sprague-Dawley , Uniones Estrechas/ultraestructura
7.
J Stroke Cerebrovasc Dis ; 30(6): 105760, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33845422

RESUMEN

Dentin matrix protein 1 (DMP1) is an extracellular matrix phosphoprotein that is known to facilitate mineralization of collagen in bone and promote osteoblast/odontoblast differentiation. Blood-brain barrier (BBB) disruption is the major pathogenesis in secondary brain injury after intracerebral hemorrhage (ICH). This study aimed to investigate the expression pattern of DMP1 in the mouse brain and explore the role of DMP1 in BBB disruption and brain injury in a mouse model of ICH. Mice were subjected to autologous blood injection-induced ICH. Immunofluorescence staining, western blot analysis, neurobehavioral tests, brain water content measurements, Evans blue permeability assay, and transmission electron microscopy were performed. Small interfering RNA targeting DMP1 (DMP1 siRNA) was administered at 72 h prior to ICH. Results showed that DMP1 is expressed extensively in the mouse brain, and is upregulated in the ICH model. Administration of DMP1 siRNA effectively ameliorated BBB disruption, attenuated brain edema, and improved neurological function after ICH. Moreover, the expression of zonula occludens-1 (ZO-1) and occludin were upregulated, and matrix metalloproteinase-9 (MMP-9) was downregulated in the ICH model. DMP1 siRNA administration reversed the expression of ZO-1, occludin, and MMP-9. These results demonstrated that DMP1 upregulation plays an essential role in inducing BBB disruption and brain injury after ICH. The inhibition of DMP1 could be a potential therapeutic strategy for ICH treatment.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Edema Encefálico/prevención & control , Hemorragia Cerebral/terapia , Proteínas de la Matriz Extracelular/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Animales , Barrera Hematoencefálica/ultraestructura , Edema Encefálico/genética , Edema Encefálico/metabolismo , Edema Encefálico/patología , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ocludina/genética , Ocludina/metabolismo , ARN Interferente Pequeño/genética , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
8.
Microvasc Res ; 132: 104054, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32768464

RESUMEN

Ischemic stroke is a leading cause of mortality worldwide that occurs following the reduction or interruption of blood brain supply, characterized by a cascade of early events as oxidative stress and ensuing neuro-inflammation, energy failure and the burst of intracellular Ca++ resulting in activation of phospholipases and large increase in FFA including arachidonic acid, ultimately leading to nervous cell death. Grape Seed Flour (GSF) is a complex polyphenolic mixture harboring antioxidant, anti-inflammatory and neuroprotective properties. Orlistat (Xenical ™,Xe) is a gastro-intestinal lipase inhibitor and an anti-obesity agent. In an earlier study we reported the higher efficiency in neuroprotection against HFD-induced brain lipotoxicity when combining the two drugs (GSF + Xe). As a result repurposing Xe as an adjunct to GSF therapy against stroke appeared relevant and worthy of investigation. I/R insult disrupted the blood brain barrier (BBB) as assessed by EB dye extravasation, increased water and Na+ within the brain. Ultrastructurally I/R altered the brain blood capillaries at the vicinity of hippocampus dentate gyrus area as assessed by transmission and scanning electron microscopy. I/R altered lipid metabolism as revealed by LDL/HDL ratio, lipase activity, and FFA profiles. Moreover, I/R induced neuro-inflammation as assessed by down-regulation of anti-inflammatory CD 56 and up-regulation of pro-inflammatory CD 68 antigen. Importantly almost all I/R-induced disturbances were retrieved partially upon Xe or GSF on their own, and optimally when combining the two drugs. Xe per se is protective against I/R injury and the best neuroprotection was obtained when associating low dosage Xe with high dosage GSF, enabling neuroprevention and cell survival within hippocampus dentate gyrus area as revealed by increased staining of Ki 67 proliferation biomarker.


Asunto(s)
Antiinflamatorios/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/prevención & control , Permeabilidad Capilar/efectos de los fármacos , Extracto de Semillas de Uva/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Reguladores del Metabolismo de Lípidos/farmacología , Fármacos Neuroprotectores/farmacología , Orlistat/farmacología , Daño por Reperfusión/prevención & control , Accidente Cerebrovascular/prevención & control , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Edema Encefálico/metabolismo , Edema Encefálico/patología , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Masculino , Ratas Wistar , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
9.
J Cell Mol Med ; 23(2): 819-827, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30450815

RESUMEN

The blood-brain barrier (BBB) is essential for a functional neurovascular unit. Most studies focused on the cells forming the BBB, but very few studied the basement membrane (BM) of brain capillaries in ageing. We used transmission electron microscopy and electron tomography to investigate the BM of the BBB in ageing C57BL/6J mice. The thickness of the BM of the BBB from 24-month-old mice was double as compared with that of 6-month-old mice (107 nm vs 56 nm). The aged BBB showed lipid droplets gathering within the BM which further increased its thickness (up to 572 nm) and altered its structure. The lipids appeared to accumulate toward the glial side of the BM. Electron tomography showed that the lipid-rich BM regions are located in small pockets formed by the end-feet of astrocytes. These findings suggest an imbalance of the lipid metabolism and that may precede the structural alteration of the BM. These alterations may favour the accretion of abnormal proteins that lead to neurodegeneration in ageing. These findings warrant further investigation of the BM of brain capillaries and of adjoining cells as potential targets for future therapies.


Asunto(s)
Envejecimiento/fisiología , Membrana Basal/ultraestructura , Barrera Hematoencefálica/ultraestructura , Capilares/ultraestructura , Músculo Liso Vascular/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Animales , Astrocitos/metabolismo , Astrocitos/ultraestructura , Membrana Basal/metabolismo , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/ultraestructura , Capilares/metabolismo , Tomografía con Microscopio Electrónico , Gotas Lipídicas/metabolismo , Gotas Lipídicas/ultraestructura , Metabolismo de los Lípidos/fisiología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neuroglía/metabolismo , Neuroglía/ultraestructura
10.
Biochim Biophys Acta Mol Cell Res ; 1864(8): 1382-1392, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28495656

RESUMEN

Tight junctions (TJ) between brain endothelial cells are essential for formation and maintenance of the blood-brain barrier (BBB). Although loss of BBB integrity is associated with several neuropathological disorders, treatments that augment or stabilise the BBB are scarce. Here we show that physiological concentrations of dehydroepiandrosterone sulfate (DHEAS) stimulate the expression of the TJ proteins zonula occludens-1 (ZO-1) and claudin-3 in the brain-derived endothelial cell line bEnd.3 and promote TJ formation between neighbouring cells, demonstrated by augmented transendothelial resistance across cell monolayers. Silencing androgen receptor expression by siRNA does not prevent DHEAS-induced stimulation of ZO-1 expression, indicating that conversion of DHEAS into testosterone is not required for its actions. Suppression of Gnα11 expression by siRNA prevents DHEAS actions, pointing towards a G-protein-coupled receptor as being a mediator of the DHEAS effects. These results are consistent with the idea that DHEAS, acting as a hormone in its own right, supports the integrity of the BBB. The current findings might help in developing new strategies for the prevention or treatment of neurological disorders associated with BBB defects.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Sulfato de Deshidroepiandrosterona/farmacología , Células Endoteliales/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Proteína de la Zonula Occludens-1/genética , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Línea Celular , Claudina-3/genética , Claudina-3/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Subunidades alfa de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Ratones , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Proteína de la Zonula Occludens-1/agonistas , Proteína de la Zonula Occludens-1/metabolismo
11.
Glia ; 66(1): 126-144, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28925083

RESUMEN

Stimulation of Na+ /H+ exchanger isoform 1 (NHE1) in astrocytes causes ionic dysregulation under ischemic conditions. In this study, we created a Nhe1flox/flox (Nhe1f/f ) mouse line with exon 5 of Nhe1 flanked with two loxP sites and selective ablation of Nhe1 in astrocytes was achieved by crossing Nhe1f/f mice with Gfap-CreERT2 Cre-recombinase mice. Gfap-CreERT2+/- ;Nhe1f/f mice at postnatal day 60-90 were treated with either corn oil or tamoxifen (Tam, 75 mg/kg/day, i.p.) for 5 days. After 30 days post-injection, mice underwent transient middle cerebral artery occlusion (tMCAO) to induce ischemic stroke. Compared with the oil-vehicle group (control), Tam-treated Gfap-CreERT2+/- ;Nhe1f/f (Nhe1 KO) mice developed significantly smaller ischemic infarction, less edema, and less neurological function deficits at 1-5 days after tMCAO. Immunocytochemical analysis revealed less astrocytic proliferation, less cellular hypertrophy, and less peri-lesion gliosis in Nhe1 KO mouse brains. Selective deletion of Nhe1 in astrocytes also reduced cerebral microvessel damage and blood-brain barrier (BBB) injury in ischemic brains. The BBB microvessels of the control brains show swollen endothelial cells, opened tight junctions, increased expression of proinflammatory protease MMP-9, and significant loss of tight junction protein occludin. In contrast, the Nhe1 KO mice exhibited reduced BBB breakdown and normal tight junction structure, with increased expression of occludin and reduced MMP-9. Most importantly, deletion of astrocytic Nhe1 gene significantly increased regional cerebral blood flow in the ischemic hemisphere at 24 hr post-MCAO. Taken together, our study provides the first line of evidence for a causative role of astrocytic NHE1 protein in reactive astrogliosis and ischemic neurovascular damage.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/patología , Gliosis/patología , Infarto de la Arteria Cerebral Media/complicaciones , Intercambiador 1 de Sodio-Hidrógeno/deficiencia , Animales , Astrocitos/ultraestructura , Barrera Hematoencefálica/ultraestructura , Infarto Encefálico/diagnóstico , Infarto Encefálico/etiología , Infarto Encefálico/genética , Circulación Cerebrovascular/genética , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/etiología , Gliosis/genética , Gliosis/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Actividad Motora/genética , Examen Neurológico , Reperfusión , Intercambiador 1 de Sodio-Hidrógeno/genética
12.
J Neuroinflammation ; 15(1): 50, 2018 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-29463289

RESUMEN

BACKGROUND: Echovirus (E) 30 (E-30) meningitis is characterized by neuroinflammation involving immune cell pleocytosis at the protective barriers of the central nervous system (CNS). In this context, infection of the blood-cerebrospinal fluid barrier (BCSFB), which has been demonstrated to be involved in enteroviral CNS pathogenesis, may affect the tight junction (TJ) and adherens junction (AJ) function and morphology. METHODS: We used an in vitro human choroid plexus epithelial (HIBCPP) cell model to investigate the effect of three clinical outbreak strains (13-311, 13-759, and 14-397) isolated in Germany in 2013, and compared them to E-30 Bastianni. Conducting transepithelial electrical resistance (TEER), paracellular dextran flux measurement, quantitative real-time polymerase chain reaction (qPCR), western blot, and immunofluorescence analysis, we investigated TJ and AJ function and morphology as well as strain-specific E-30 infection patterns. Additionally, transmission electron and focused ion beam microscopy electron microscopy (FIB-SEM) was used to evaluate the mode of leukocyte transmigration. Genome sequencing and phylogenetic analyses were performed to discriminate potential genetic differences among the outbreak strains. RESULTS: We observed a significant strain-dependent decrease in TEER with strains E-30 Bastianni and 13-311, whereas paracellular dextran flux was only affected by E-30 Bastianni. Despite strong similarities among the outbreak strains in replication characteristics and particle distribution, strain 13-311 was the only outbreak isolate revealing comparable disruptive effects on TJ (Zonula Occludens (ZO) 1 and occludin) and AJ (E-cadherin) morphology to E-30 Bastianni. Notwithstanding significant junctional alterations upon E-30 infection, we observed both para- and transcellular leukocyte migration across HIBCPP cells. Complete genome sequencing revealed differences between the strains analyzed, but no explicit correlation with the observed strain-dependent effects on HIBCPP cells was possible. CONCLUSION: The findings revealed distinct E-30 strain-specific effects on barrier integrity and junctional morphology. Despite E-30-induced barrier alterations leukocyte trafficking did not exclusively occur via the paracellular route.


Asunto(s)
Barrera Hematoencefálica/virología , Líquido Cefalorraquídeo/virología , Plexo Coroideo/virología , Brotes de Enfermedades , Enterovirus Humano B/aislamiento & purificación , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Línea Celular Tumoral , Supervivencia Celular/fisiología , Células Cultivadas , Líquido Cefalorraquídeo/metabolismo , Plexo Coroideo/metabolismo , Plexo Coroideo/ultraestructura , Enterovirus Humano B/metabolismo , Humanos , Filogenia , Especificidad de la Especie
13.
Stem Cells ; 35(5): 1246-1258, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28142208

RESUMEN

Stroke is a life-threatening disease with limited therapeutic options. Cell therapy has emerged as an experimental stroke treatment. Blood-brain barrier (BBB) impairment is a key pathological manifestation of ischemic stroke, and barrier repair is an innovative target for neurorestoration in stroke. Here, we evaluated via electron microscopy the ability of transplanted human bone marrow endothelial progenitor cells (hBMEPCs) to repair the BBB in adult Sprague-Dawley rats subjected to transient middle cerebral artery occlusion (tMCAO). ß-galactosidase prelabeled hBMEPCs were intravenously transplanted 48 hours post-tMCAO. Ultrastructural analysis of microvessels in nontransplant stroke rats revealed typical BBB pathology. At 5 days post-transplantation with hBMEPCs, stroke rats displayed widespread vascular repair in bilateral striatum and motor cortex, characterized by robust cell engraftment within capillaries. hBMEPC transplanted stroke rats exhibited near normal morphology of endothelial cells (ECs), pericytes, and astrocytes, without detectable perivascular edema. Near normal morphology of mitochondria was also detected in ECs and perivascular astrocytes from transplanted stroke rats. Equally notable, we observed numerous pinocytic vesicles within engrafted cells. Robust engraftment and intricate functionality of transplanted hBMEPCs likely abrogated stroke-altered vasculature. Preserving mitochondria and augmenting pinocytosis in cell-based therapeutics represent a new neurorestorative mechanism in BBB repair for stroke. Stem Cells 2017;35:1246-1258.


Asunto(s)
Barrera Hematoencefálica/patología , Trasplante de Médula Ósea , Isquemia Encefálica/terapia , Capilares/patología , Células Progenitoras Endoteliales/trasplante , Mitocondrias/patología , Pinocitosis , Accidente Cerebrovascular/terapia , Administración Intravenosa , Animales , Astrocitos/patología , Astrocitos/ultraestructura , Barrera Hematoencefálica/ultraestructura , Isquemia Encefálica/complicaciones , Capilares/ultraestructura , Separación Celular , Células Progenitoras Endoteliales/citología , Humanos , Masculino , Mitocondrias/ultraestructura , Ratas Sprague-Dawley , Accidente Cerebrovascular/complicaciones , beta-Galactosidasa/metabolismo
14.
PLoS Biol ; 13(10): e1002279, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26512759

RESUMEN

Aging is the major risk factor for neurodegenerative diseases such as Alzheimer's disease, but little is known about the processes that lead to age-related decline of brain structures and function. Here we use RNA-seq in combination with high resolution histological analyses to show that aging leads to a significant deterioration of neurovascular structures including basement membrane reduction, pericyte loss, and astrocyte dysfunction. Neurovascular decline was sufficient to cause vascular leakage and correlated strongly with an increase in neuroinflammation including up-regulation of complement component C1QA in microglia/monocytes. Importantly, long-term aerobic exercise from midlife to old age prevented this age-related neurovascular decline, reduced C1QA+ microglia/monocytes, and increased synaptic plasticity and overall behavioral capabilities of aged mice. Concomitant with age-related neurovascular decline and complement activation, astrocytic Apoe dramatically decreased in aged mice, a decrease that was prevented by exercise. Given the role of APOE in maintaining the neurovascular unit and as an anti-inflammatory molecule, this suggests a possible link between astrocytic Apoe, age-related neurovascular dysfunction and microglia/monocyte activation. To test this, Apoe-deficient mice were exercised from midlife to old age and in contrast to wild-type (Apoe-sufficient) mice, exercise had little to no effect on age-related neurovascular decline or microglia/monocyte activation in the absence of APOE. Collectively, our data shows that neurovascular structures decline with age, a process that we propose to be intimately linked to complement activation in microglia/monocytes. Exercise prevents these changes, but not in the absence of APOE, opening up new avenues for understanding the complex interactions between neurovascular and neuroinflammatory responses in aging and neurodegenerative diseases such as Alzheimer's disease.


Asunto(s)
Envejecimiento , Apolipoproteínas E/metabolismo , Astrocitos/metabolismo , Complemento C1q/metabolismo , Actividad Motora , Enfermedades Neurodegenerativas/prevención & control , Enfermedades Vasculares/prevención & control , Animales , Apolipoproteínas E/sangre , Apolipoproteínas E/química , Apolipoproteínas E/genética , Astrocitos/inmunología , Astrocitos/patología , Astrocitos/ultraestructura , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/ultraestructura , Activación de Complemento , Complemento C1q/genética , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica , Homocigoto , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Monocitos/ultraestructura , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Plasticidad Neuronal , Acoplamiento Neurovascular , Estabilidad Proteica , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
15.
Med Sci Monit ; 24: 880-890, 2018 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-29432410

RESUMEN

BACKGROUND Angiopoietin-like 4 (ANGPTL4) is neuroprotective when administered acutely for the treatment of cerebral ischemia. The aim of the present study was to evaluate the preventive effects of ANGPTL4 on the formation of brain edema and to determine whether it promotes the recovery of neurological function following intracerebral hemorrhage (ICH). MATERIAL AND METHODS Recombinant human ANGPTL4 (rhANGPTL4; 40 µg/kg) or a vehicle was administered intraperitoneally 5 min prior to bacterial collagenase-induced ICH in male C57/B6J mice. Behavioral tests were performed prior to ICH and at days 1, 3, 7, 14, 21, and 28 after ICH. Brain edema and hematoma volume were examined separately using the wet weight/dry weight method and hematoxylin-eosin staining. The integrity of the tight and adherens junctions was quantified via immunofluorescence. The ultrastructure of the blood-brain barrier (BBB) was examined using transmission electron microscopy. Vascular endothelial (VE)-cadherin, claudin-5, Src, and phospho-Src in the ipsilateral and contralateral striatum were detected by Western blot analysis. RESULTS RhANGPTL4 reduced brain edema and hematoma volume and improved neurological functional recovery over the subsequent 4 weeks when compared with the control group. rhANGPTL4 significantly increased VE-cadherin and claudin-5-positive areas and relative amounts in the peri­hematoma region compared with the control group. In addition, ANGPTL4 significantly reduced the ratio of phospho-Src to Src. The significant reduction of Src kinase activity in the peri­hematoma region of ANGPTL-treated mice was paralleled by a decrease in vascular permeability and edema formation. CONCLUSIONS These results suggest that ANGPTL4 is a relevant target for vasculoprotection and cerebral protection during stroke.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/uso terapéutico , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/tratamiento farmacológico , Proteína 4 Similar a la Angiopoyetina/farmacología , Animales , Antígenos CD/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/ultraestructura , Edema Encefálico/patología , Cadherinas/metabolismo , Hemorragia Cerebral/patología , Claudina-5/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Hematoma/complicaciones , Hematoma/tratamiento farmacológico , Hematoma/patología , Humanos , Masculino , Ratones Endogámicos C57BL
16.
Neurobiol Dis ; 103: 123-132, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28365214

RESUMEN

The neuronal ceroid lipofuscinoses are a group of recessively inherited, childhood-onset neurodegenerative conditions. Several forms are caused by mutations in genes encoding putative lysosomal membrane proteins. Studies of the cell biology underpinning these disorders are hampered by the poor antigenicity of the membrane proteins, which makes visualization of the endogenous proteins difficult. We have used Drosophila to generate knock-in YFP-fusions for two of the NCL membrane proteins: CLN7 and CLN3. The YFP-fusions are expressed at endogenous levels and the proteins can be visualized live without the need for overexpression. Unexpectedly, both CLN7 and CLN3 have restricted expression in the CNS of Drosophila larva and are predominantly expressed in the glia that form the insect blood-brain-barrier. CLN7 is also expressed in neurons in the developing visual system. Analogous with murine CLN3, Drosophila CLN3 is strongly expressed in the excretory and osmoregulatory Malpighian tubules, but the knock-in also reveals unexpected localization of the protein to the apical domain adjacent to the lumen. In addition, some CLN3 protein in the tubules is localized within mitochondria. Our in vivo imaging of CLN7 and CLN3 suggests new possibilities for function and promotes new ideas about the cell biology of the NCLs.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas de Drosophila/biosíntesis , Túbulos de Malpighi/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de Transporte de Membrana/biosíntesis , Lipofuscinosis Ceroideas Neuronales/metabolismo , Neuronas/metabolismo , Animales , Barrera Hematoencefálica/química , Barrera Hematoencefálica/ultraestructura , Drosophila , Proteínas de Drosophila/análisis , Proteínas de Drosophila/genética , Expresión Génica , Técnicas de Sustitución del Gen , Túbulos de Malpighi/química , Túbulos de Malpighi/ultraestructura , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/análisis , Proteínas de Transporte de Membrana/genética , Lipofuscinosis Ceroideas Neuronales/genética , Neuronas/química , Neuronas/ultraestructura
17.
Histochem Cell Biol ; 148(1): 3-12, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28283744

RESUMEN

Due to the physical and physiological properties of the blood-brain barrier (BBB), the transport of neurotherapeutics from blood to brain is still a pharmaceutical challenge. We previously conducted a series of experiments to explore the potential of the anti-transferrin receptor 8D3 monoclonal antibody (mAb) to transport neurotherapeutics across the BBB. In that study, gold nanoparticles (AuNPs) were coated with the 8D3 antibody and administered intravenously to mice. Transmission electron microscopy was used and a two-dimensional (2D) image analysis was performed to detect the AuNPs in the brain capillary endothelial cells (BCECs) and brain parenchyma. In the present work, we determined that serial block-face scanning electron microscopy (SBF-SEM) is a useful tool to study the transcytosis of these AuNPs across the BBB in three dimensions and we, therefore, applied it to gain more knowledge of their transcellular trafficking. The resulting 3D reconstructions provided additional information on the endocytic vesicles containing AuNPs and the endosomal processing that occurs inside BCECs. The passage from 2D to 3D analysis reinforced the trafficking model proposed in the 2D study, and revealed that the vesicles containing AuNPs are significantly larger and more complex than described in our 2D study. We also discuss tradeoffs of using this technique for our application, and conclude that together with other volume electron microscopy imaging techniques, SBF-SEM is a powerful approach that is worth of considering for studies of drug transport across the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Oro/farmacocinética , Nanopartículas del Metal/análisis , Microscopía Electrónica de Rastreo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/farmacocinética , Oro/administración & dosificación , Inyecciones Intravenosas , Masculino , Nanopartículas del Metal/administración & dosificación , Ratones , Ratones Endogámicos ICR
18.
Cerebrovasc Dis ; 44(1-2): 10-25, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28402976

RESUMEN

BACKGROUND: Apelin-13 has been found to have protective effects on many neurological diseases, including cerebral ischemia. However, whether Apelin-13 acts on blood-brain barrier (BBB) disruption following cerebral ischemia is largely unknown. Aquaporin-4 (AQP4) has a close link with BBB due to the high concentration in astrocyte foot processes and regulation of astrocytes function. Here, we aimed to test Apelin-13's effects on ischemic BBB injury and examine whether the effects were dependent on AQP4. METHODS: We detected the expression of AQP4 induced by Apelin-13 injection at 1, 3, and 7 days after middle cerebral artery occlusion. Meanwhile, we examined the effects of Apelin-13 on neurological function, infarct volume, and BBB disruption owing to cerebral ischemia in wild type mice, and tested whether such effects were AQP4 dependent by using AQP4 knock-out mice. Furthermore, we assessed the possible signal transduction pathways activated by Apelin-13 to regulate AQP4 expression via astrocyte cultures. RESULTS: It was found that Apelin-13 highly increased AQP4 expression as well as reduced neurological scores and infarct volume. Importantly, Apelin-13 played a role of BBB protection in both types of mice by reducing BBB permeability, increased vascular endothelial growth factor, upregulated endothelial nitric oxide synthase, and downregulated inducible NOS. In morphology, we demonstrated Apelin-13 suppressed tight junction opening and endothelial cell swelling via electron microscopy detection. Meanwhile, Apelin-13 also alleviated apoptosis of astrocytes and promoted angiogenesis. Interestingly, effects of AQP4 on neurological function and infarct volume varied with time course, while AQP4 elicited protective effects on BBB at all time points. Statistical analysis of 2-way analysis of variance with replication indicated that AQP4 was required for these effects. In addition, Apelin-13 upregulated phosphorylation of extracellular signal-regulated kinase (ERK) and Akt as well as AQP4 protein in cultured astrocytes. The latter was inhibited by ERK and phosphatidylinositol 3'-kinase (PI3K) inhibitors. CONCLUSION: Our data suggest that Apelin-13 protects BBB from disruption after cerebral ischemia both morphologically and functionally, which is highly associated with the increased levels of AQP4, possibly through the activation of ERK and PI3K/Akt pathways. This study provides double targets to protection of ischemic BBB damage, which can present new insights to drugs development.


Asunto(s)
Acuaporina 4/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intercelular/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Acuaporina 4/deficiencia , Acuaporina 4/genética , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Barrera Hematoencefálica/ultraestructura , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Exp Parasitol ; 174: 31-41, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28011167

RESUMEN

Trypanosoma brucei are extracellular hemoflagellate protozoan parasites and one of the causative agents of a devastating zoonotic disease called African Trypanosomiasis. In humans, the disease is caused by Trypanosoma brucei rhodensiense and Trypanosoma brucei gambiense, which cross the blood brain barrier (BBB) causing neurological disorders which culminate in death if untreated. In some domestic animals and laboratory rodents, Trypanosoma brucei brucei causes a disease similar to that in humans. The mechanism by which Trypanosoma brucei brucei invade biological barriers including the BBB has not been fully elucidated. To further address this issue, Mardin Dardy Canine Kidney II (MDCKII) and Human dermal microvascular endothelial cell (HDMEC) monolayers were grown to confluence on transwell inserts to constitute in vitro biological barriers. MDCKII cells were chosen for their ability to form tight junctions similar to those formed by the BBB endothelial cells. Labeled trypanosomes were placed in the upper chamber of transwell inserts layered with confluent MDCKII/HDMEC monolayers and their ability to cross the monolayer over time evaluated. Our results show that only 0.5-1.25% of Trypanosoma brucei brucei were able to migrate across the monolayers after 3 h. By employing immune-staining and confocal microscopic analysis we observed that trypanosomes were located at the tight junctions and inside the cell in the MDCK II monolayers indicating that they crossed the monolayer using both the paracellular and transcellular routes. Our observations also showed that there seemed to be no obvious degradation of junction proteins Zonula Ocludens-1, Occludin and Ecadherin. In the HDMEC cell monolayer, our scanning electron microscopy data showed that Trypanosoma brucei brucei is able to modulate the plasma membrane to form invaginations similar to cuplike structures formed by Tlymphocytes. However these structures seemed to be independent of vascular adhesion molecules suggesting that they could be more like the membrane ruffles formed by certain intracellular bacteria during invasion. Taken together, our data reveal a mechanism by which Trypanosoma brucei brucei is able to cross different biological barriers including the BBB without causing any obvious damage.


Asunto(s)
Barrera Hematoencefálica/parasitología , Células de Riñón Canino Madin Darby/parasitología , Trypanosoma brucei brucei/fisiología , Tripanosomiasis Africana/parasitología , Animales , Barrera Hematoencefálica/ultraestructura , Línea Celular , Membrana Celular/parasitología , Membrana Celular/ultraestructura , Perros , Flagelos/fisiología , Flagelos/ultraestructura , Técnica del Anticuerpo Fluorescente , Humanos , Células de Riñón Canino Madin Darby/ultraestructura , Ratones , Microscopía Confocal , Microscopía Electrónica de Rastreo , Conejos , Ratas , Proteínas de Uniones Estrechas/química , Uniones Estrechas/parasitología , Trypanosoma brucei brucei/ultraestructura , Tripanosomiasis Africana/patología
20.
J Stroke Cerebrovasc Dis ; 26(10): 2065-2073, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28669655

RESUMEN

OBJECTIVE: IMM-H004 (7-hydroxy-5-methoxy-4-methyl-3-[4-methylpiperazin-1-yl]-2H-chromen-2-one) is a novel coumarin derivative that showed better effect in improving global cerebral ischemia in rats. However, the effects and mechanisms in focal cerebral ischemia were not clear. Blood-brain barrier (BBB) protection is a vital strategy for the treatment of cerebral ischemia. This study is to investigate whether IMM-H004 improves brain ischemia injury via BBB protection. METHODS: Focal brain ischemia model was induced by middle cerebral artery occlusion for 1 hour and reperfusion (MCAO/R) for 24 hours in rats. IMM-H004 (1.5, 3, 6 mg/kg) and edaravone (positive drug, 6 mg/kg) were administered after 5 minutes of occlusion. Neurological score and TTC staining were used to evaluate the effect of IMM-H004. Evans Blue (EB) staining and electron microscopy were used to assess BBB permeability. Western blot, reverse transcription-polymerase chain reaction, and immunohistochemistry were used to detect the expression of BBB structure-related proteins. RESULTS: Compared with the model group, IMM-H004 in the focal brain ischemia model improved neurological function and reduced cerebral infarction size and edema content. IMM-H004 sharply reduced the EB content and alleviated BBB structure. In addition, IMM-H004 increased the level of zonula occludens (ZO-1) and occluding, decreased the level of aquaporin 4 and matrix metalloproteinase 9, either in cortex or in hippocampus. And all of these changed were related to BBB protection. CONCLUSION: IMM-H004 improved cerebral ischemia injury via BBB protection. For a potential therapy drug of cerebral ischemia, IMM-H004 merits further study.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Cumarinas/farmacología , Infarto de la Arteria Cerebral Media/prevención & control , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/prevención & control , Animales , Acuaporina 4/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/ultraestructura , Edema Encefálico/metabolismo , Edema Encefálico/patología , Edema Encefálico/prevención & control , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Citoprotección , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ocludina/metabolismo , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factores de Tiempo , Proteína de la Zonula Occludens-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA