Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Biomolecules ; 11(12)2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34944506

RESUMO

Renin-angiotensin systems produce angiotensin II (Ang II) and angiotensin 1-7 (Ang 1-7), which are able to induce opposite effects on circulation. This study in vivo assessed the effects induced by Ang II or Ang 1-7 on rat pial microcirculation during hypoperfusion-reperfusion, clarifying the mechanisms causing the imbalance between Ang II and Ang 1-7. The fluorescence microscopy was used to quantify the microvascular parameters. Hypoperfusion and reperfusion caused vasoconstriction, disruption of blood-brain barrier, reduction of capillary perfusion and an increase in reactive oxygen species production. Rats treated with Ang II showed exacerbated microvascular damage with stronger vasoconstriction compared to hypoperfused rats, a further increase in leakage, higher decrease in capillary perfusion and marker oxidative stress. Candesartan cilexetil (specific Ang II type 1 receptor (AT1R) antagonist) administration prior to Ang II prevented the effects induced by Ang II, blunting the hypoperfusion-reperfusion injury. Ang 1-7 or ACE2 activator administration, preserved the pial microcirculation from hypoperfusion-reperfusion damage. These effects of Ang 1-7 were blunted by a Mas (Mas oncogene-encoded protein) receptor antagonist, while Ang II type 2 receptor antagonists did not affect Ang 1-7-induced changes. In conclusion, Ang II and Ang 1-7 triggered different mechanisms through AT1R or MAS receptors able to affect cerebral microvascular injury.


Assuntos
Angiotensina II/administração & dosagem , Angiotensina I/administração & dosagem , Benzimidazóis/administração & dosagem , Compostos de Bifenilo/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Pia-Máter/irrigação sanguínea , Traumatismo por Reperfusão/metabolismo , Tetrazóis/administração & dosagem , Angiotensina I/efeitos adversos , Angiotensina II/efeitos adversos , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo/farmacologia , Feminino , Masculino , Microcirculação/efeitos dos fármacos , Microscopia de Fluorescência , Fragmentos de Peptídeos/efeitos adversos , Pia-Máter/efeitos dos fármacos , Pia-Máter/metabolismo , Proto-Oncogene Mas/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Tetrazóis/farmacologia
2.
Cells ; 10(1)2020 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-33375610

RESUMO

Although del Río-Hortega originally reported that leptomeningeal cells are the source of ramified microglia in the developing brain, recent views do not seem to pay much attention to this notion. In this study, in vitro experiments were conducted to determine whether leptomeninges generate ramified microglia. The leptomeninges of neonatal rats containing Iba1+ macrophages were peeled off the brain surface. Leptomeningeal macrophages strongly expressed CD68 and CD163, but microglia in the brain parenchyma did not. Leptomeningeal macrophages expressed epidermal growth factor receptor (EGFR) as revealed by RT-PCR and immunohistochemical staining. Cells obtained from the peeled-off leptomeninges were cultured in a serum-free medium containing EGF, resulting in the formation of large cell aggregates in which many proliferating macrophages were present. In contrast, colony-stimulating factor 1 (CSF1) did not enhance the generation of Iba1+ cells from the leptomeningeal culture. The cell aggregates generated ramified Iba1+ cells in the presence of serum, which express CD68 and CD163 at much lower levels than primary microglia isolated from a mixed glial culture. Therefore, the leptomeningeal-derived cells resembled parenchymal microglia better than primary microglia. This study suggests that microglial progenitors expressing EGFR reside in the leptomeninges and that there is a population of microglia-like cells that grow independently of CSF1.


Assuntos
Macrófagos , Microglia , Pia-Máter , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular , Células Cultivadas , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microglia/citologia , Microglia/metabolismo , Pia-Máter/citologia , Pia-Máter/metabolismo , Ratos , Ratos Wistar , Receptores de Superfície Celular/metabolismo
3.
Cephalalgia ; 40(12): 1310-1320, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32611244

RESUMO

BACKGROUND: The Transient Receptor Potential Ankyrin 1 (TRPA1) channel might play a role in migraine. However, different mechanisms for this have been suggested. The purpose of our study was to investigate the localization and significance of TRPA1 channels in rat pial and dural arteries. METHODS: Immunofluorescence microscopy was used to localize TRPA1 channels in dural arteries, pial arteries, dura mater and trigeminal ganglion. The genuine closed cranial window model was used to examine the effect of Na2S, a donor of the TRPA1 channel opener H2S, on the diameter of pial and dural arteries. Further, we performed blocking experiments with TRPA1 antagonist HC-030031, calcitonin gene-related peptide (CGRP) receptor antagonist olcegepant and KCa3.1 channel blocker TRAM-34. RESULTS: TRPA1 channels were localized to the endothelium of both dural and pial arteries and in nerve fibers in dura mater. Further, we found TRPA1 expression in the membrane of trigeminal ganglia neuronal cells, some of them also staining for CGRP. Na2S caused dilation of both dural and pial arteries. In dural arteries, this was inhibited by HC-030031 and olcegepant. In pial arteries, the dilation was inhibited by TRAM-34, suggesting involvement of the KCa3.1 channel. CONCLUSION: Na2S causes a TRPA1- and CGRP-dependent dilation of dural arteries and a KCa3.1 channel-dependent dilation of pial arteries in rats.


Assuntos
Dura-Máter/metabolismo , Pia-Máter/metabolismo , Sulfetos/farmacologia , Canal de Cátion TRPA1/metabolismo , Vasodilatadores/farmacologia , Animais , Dura-Máter/efeitos dos fármacos , Masculino , Transtornos de Enxaqueca/metabolismo , Transtornos de Enxaqueca/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pia-Máter/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Canal de Cátion TRPA1/efeitos dos fármacos
4.
J Neurol ; 267(1): 239-243, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31620867

RESUMO

OBJECTIVE: Deferiprone is an iron chelator that has recently been used to treat patients with infratentorial superficial siderosis (iSS). It is considered to have a generally favourable safety profile but concerns have been raised due to the risk of agranulocytosis. We aimed to evaluate the safety and tolerability of oral deferiprone as a treatment for patients with iSS. METHODS: We present a case series of 10 consecutive patients presenting with classical iSS treated with deferiprone. RESULTS: Ten patients were followed up for a mean period of 2.3 years (range 0.5-5.5 years). Four patients (40%) were withdrawn from treatment because of treatment-related side effects. The reasons for treatment discontinuation were neutropenic sepsis (n = 3) and fatigue (n = 1). In 2 out of the 3 cases of neutropenic sepsis, patients initially developed neutropenia without sepsis. The mean time to neutropenic sepsis following deferiprone was 1.2 years (range 0.3-2.5) with mean neutrophil count of 0.4 (range 0.3-0.5). Six patients (60%) reported no change in neurological function while on treatment, and four patients (40%) reported that their condition deteriorated. CONCLUSIONS: Deferiprone was poorly tolerated, with 40% of patients withdrawing from treatment, most commonly due to neutropenic sepsis, after an average of 2 years on treatment. This study increases the number of reported cases of agranulocytosis in patients with iSS treated with deferiprone. Clinicians treating iSS patients with deferiprone should be aware that this drug has a potentially life-threatening side effect of neutropenic sepsis, and should ensure that appropriate haematological monitoring is in place.


Assuntos
Agranulocitose/induzido quimicamente , Encefalopatias/tratamento farmacológico , Deferiprona/efeitos adversos , Hemossiderose/tratamento farmacológico , Quelantes de Ferro/efeitos adversos , Pia-Máter/metabolismo , Doenças da Medula Espinal/tratamento farmacológico , Adulto , Idoso , Encefalopatias/diagnóstico por imagem , Feminino , Seguimentos , Hemossiderose/diagnóstico por imagem , Humanos , Masculino , Pessoa de Meia-Idade , Pia-Máter/diagnóstico por imagem , Doenças da Medula Espinal/diagnóstico por imagem
5.
JCI Insight ; 4(20)2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31619586

RESUMO

Intrathecal (IT) delivery and pharmacology of antisense oligonucleotides (ASOs) for the CNS have been successfully developed to treat spinal muscular atrophy. However, ASO pharmacokinetic (PK) and pharmacodynamic (PD) properties remain poorly understood in the IT compartment. We applied multimodal imaging techniques to elucidate the IT PK and PD of unlabeled, radioactively labeled, or fluorescently labeled ASOs targeting ubiquitously expressed or neuron-specific RNAs. Following lumbar IT bolus injection in rats, all ASOs spread rostrally along the neuraxis, adhered to meninges, and were partially cleared to peripheral lymph nodes and kidneys. Rapid association with the pia and arterial walls preceded passage of ASOs across the glia limitans, along arterial intramural basement membranes, and along white-matter axonal bundles. Several neuronal and glial cell types accumulated ASOs over time, with evidence of probable glial accumulation preceding neuronal uptake. IT doses of anti-GluR1 and anti-Gabra1 ASOs markedly reduced the mRNA and protein levels of their respective neurotransmitter receptor protein targets by 2 weeks and anti-Gabra1 ASOs also reduced binding of the GABAA receptor PET ligand 18F-flumazenil in the brain over 4 weeks. Our multimodal imaging approaches elucidate multiple transport routes underlying the CNS distribution, clearance, and efficacy of IT-dosed ASOs.


Assuntos
Encéfalo/metabolismo , Antagonistas de Receptores de GABA-A/farmacocinética , Atrofia Muscular Espinal/tratamento farmacológico , Oligonucleotídeos Antissenso/farmacocinética , Animais , Artérias/diagnóstico por imagem , Artérias/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/diagnóstico por imagem , Flumazenil/administração & dosagem , Flumazenil/análogos & derivados , Antagonistas de Receptores de GABA-A/administração & dosagem , Técnicas de Silenciamento de Genes , Humanos , Injeções Espinhais , Microscopia Intravital , Masculino , Terapia de Alvo Molecular/métodos , Neuroglia/metabolismo , Neurônios/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , Pia-Máter/diagnóstico por imagem , Pia-Máter/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Receptores de AMPA/análise , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/genética , Receptores de GABA-A/análise , Receptores de GABA-A/genética , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Análise Espaço-Temporal , Tionucleotídeos/administração & dosagem , Tionucleotídeos/farmacocinética , Distribuição Tecidual
6.
Genesis ; 57(5): e23288, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30801905

RESUMO

The meninges are membranous layers surrounding the central nervous system. In the head, the meninges lie between the brain and the skull, and interact closely with both during development. The cranial meninges originate from a mesenchymal sheath on the surface of the developing brain, called primary meninx, and undergo differentiation into three layers with distinct histological characteristics: the dura mater, the arachnoid mater, and the pia mater. While genetic regulation of meningeal development is still poorly understood, mouse mutants and other models with meningeal defects have demonstrated the importance of the meninges to normal development of the calvaria and the brain. For the calvaria, the interactions with the meninges are necessary for the progression of calvarial osteogenesis during early development. In later stages, the meninges control the patterning of the skull and the fate of the sutures. For the brain, the meninges regulate diverse processes including cell survival, cell migration, generation of neurons from progenitors, and vascularization. Also, the meninges serve as a stem cell niche for the brain in the postnatal life. Given these important roles of the meninges, further investigation into the molecular mechanisms underlying meningeal development can provide novel insights into the coordinated development of the head.


Assuntos
Meninges/embriologia , Meninges/metabolismo , Meninges/fisiologia , Animais , Aracnoide-Máter/embriologia , Aracnoide-Máter/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Diferenciação Celular , Biologia do Desenvolvimento/métodos , Dura-Máter/embriologia , Dura-Máter/metabolismo , Humanos , Pia-Máter/embriologia , Pia-Máter/metabolismo , Crânio/embriologia , Crânio/metabolismo
7.
Life Sci ; 201: 150-160, 2018 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-29605446

RESUMO

AIMS: Accumulating evidence supports that cerebrospinal fluid (CSF) in the subarachnoid space (SAS) could reenter the brain parenchyma via the glymphatic influx. The present study was designed to characterize the detailed pathway of subarachnoid CSF influx by using a novel CSF tracer. MAIN METHODS: Fluorescently conjugated cadaverine (A488-ca), for the first time, was employed to investigate CSF movement in the brain. Following intracisternal infusion of CSF tracers, mice brain was sliced and prepared for fluorescence imaging. Some brain sections were immunostained in order to observe tracer distribution and cellular uptake. KEY FINDINGS: A488-ca moved into the brain parenchyma rapidly, and the influx was time and region dependent. A488-ca entered the mice brain more readily and spread more widely than another commonly used CSF tracer-fluorescently conjugated ovalbumin (OA-45). Furthermore, A488-ca could enter the brain parenchyma either along the paravascular space or across the pial surface. Suppression of glymphatic transport by administration with acetazolamide strikingly reduced the influx of A488-ca. More importantly, relative to OA-45 largely remained in the extracellular space, A488-ca exhibited obvious cellular uptake by astrocytes surrounding the blood vessels and neurons in the cerebral cortex. SIGNIFICANCE: Subarachnoid CSF could flow into the brain parenchyma via the glymphatic influx, in which the transcellular pathway was faithfully traced by intracisternal infusion with fluorescently conjugated cadaverine. These observations extend our comprehension on the glymphatic influx pathway.


Assuntos
Cadaverina/farmacologia , Líquido Cefalorraquidiano/metabolismo , Linfa/fisiologia , Espaço Subaracnóideo/metabolismo , Acetazolamida/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cadaverina/administração & dosagem , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Líquido Cefalorraquidiano/efeitos dos fármacos , Cisterna Magna , Diuréticos/farmacologia , Corantes Fluorescentes , Injeções , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pia-Máter/metabolismo , Espaço Subaracnóideo/efeitos dos fármacos
8.
Stroke ; 49(5): 1267-1275, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29669868

RESUMO

BACKGROUND AND PURPOSE: Reperfusion is the most significant determinant of good outcome after ischemic stroke. However, complete reperfusion often cannot be achieved, despite satisfactory recanalization. We hypothesized that microvascular protection was essential for achieving effective reperfusion and, hence, neuroprotection. To test this hypothesis, we have developed an in vivo model to differentially monitor parenchymal and vascular reactive oxygen species (ROS) formation. By comparing the ROS-suppressing effect of N-tert-butyl-α-phenylnitrone (PBN) with its blood-brain barrier impermeable analog 2-sulfo-phenyl-N-tert-butylnitrone (S-PBN), we assessed the impact of vascular ROS suppression alone on reperfusion and stroke outcome after recanalization. METHODS: The distal middle cerebral artery was occluded for 1 hour by compressing with a micropipette and then recanalized (n=60 Swiss mice). ROS formation was monitored for 1 hour after recanalization by intravital fluorescence microscopy in pial vasculature and cortical parenchyma with topically applied hydroethidine through a cranial window. PBN (100 mg/kg) or S-PBN (156 mg/kg) was administered shortly before recanalization, and suppression of the vascular and parenchymal hydroethidine fluorescence was examined (n=22). Microcirculatory patency, reperfusion, ischemic tissue size, and neurological outcome were also assessed in a separate group of mice 1 to 72 hours after recanalization (n=30). RESULTS: PBN and S-PBN completely suppressed the reperfusion-induced increase in ROS signal within vasculature. PBN readily suppressed ROS produced in parenchyma by 88%. S-PBN also suppressed the parenchymal ROS by 64% but starting 40 minutes later. Intriguingly, PBN and S-PBN comparably reduced the size of ischemic area by 65% and 48% (P>0.05), respectively. S-PBN restored the microvascular patency and perfusion after recanalization, suggesting that its delayed parenchymal antioxidant effect could be secondary to improved microcirculatory reperfusion. CONCLUSIONS: Promoting microvascular reperfusion by protecting vasculature can secondarily reduce parenchymal ROS formation and provide neuroprotection. The model presented can be used to directly assess pharmacological end points postulated in brain parenchyma and vasculature in vivo.


Assuntos
Benzenossulfonatos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , Microcirculação/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Pia-Máter/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Barreira Hematoencefálica , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Corantes Fluorescentes , Infarto da Artéria Cerebral Média/patologia , Microscopia Intravital , Masculino , Camundongos , Microscopia de Fluorescência , Fenantridinas , Pia-Máter/irrigação sanguínea , Pia-Máter/metabolismo , Pia-Máter/patologia , Reperfusão
9.
J Cereb Blood Flow Metab ; 38(4): 669-686, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29283289

RESUMO

Perivascular compartments surrounding central nervous system (CNS) vessels have been proposed to serve key roles in facilitating cerebrospinal fluid flow into the brain, CNS waste transfer, and immune cell trafficking. Traditionally, these compartments were identified by electron microscopy with limited molecular characterization. Using cellular markers and knowledge on cellular sources of basement membrane laminins, we here describe molecularly distinct compartments surrounding different vessel types and provide a comprehensive characterization of the arachnoid and pial compartments and their connection to CNS vessels and perivascular pathways. We show that differential expression of plectin, E-cadherin and laminins α1, α2, and α5 distinguishes pial and arachnoid layers at the brain surface, while endothelial and smooth muscle laminins α4 and α5 and smooth muscle actin differentiate between arterioles and venules. Tracer studies reveal that interconnected perivascular compartments exist from arterioles through to veins, potentially providing a route for fluid flow as well as the transport of large and small molecules.


Assuntos
Vasos Sanguíneos/fisiologia , Encéfalo/fisiologia , Líquido Cefalorraquidiano/fisiologia , Animais , Aracnoide-Máter/anatomia & histologia , Aracnoide-Máter/metabolismo , Arteríolas/metabolismo , Membrana Basal/metabolismo , Transporte Biológico , Células Endoteliais/metabolismo , Feminino , Imunidade Celular , Laminina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/metabolismo , Pia-Máter/metabolismo , Vênulas/metabolismo
10.
Cell Mol Life Sci ; 75(6): 1027-1041, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29018869

RESUMO

Originating from ectodermal epithelium, radial glial cells (RGCs) retain apico-basolateral polarity and comprise a pseudostratified epithelial layer in the developing cerebral cortex. The apical endfeet of the RGCs faces the fluid-filled ventricles, while the basal processes extend across the entire cortical span towards the pial surface. RGC functions are largely dependent on this polarized structure and the molecular components that define it. In this review, we will dissect existing molecular evidence on RGC polarity establishment and during cerebral cortex development and provide our perspective on the remaining key questions.


Assuntos
Polaridade Celular , Córtex Cerebral/metabolismo , Ectoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Neuroglia/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Junções Aderentes/metabolismo , Junções Aderentes/ultraestrutura , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/crescimento & desenvolvimento , Ventrículos Cerebrais/metabolismo , Ectoderma/citologia , Ectoderma/crescimento & desenvolvimento , Embrião de Mamíferos , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Humanos , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/citologia , Pia-Máter/citologia , Pia-Máter/crescimento & desenvolvimento , Pia-Máter/metabolismo
11.
Acta Neuropathol ; 133(4): 597-612, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28184993

RESUMO

Pathogenic autoantibodies associated with neuromyelitis optica (NMO) induce disease by targeting aquaporin-4 (AQP4) water channels enriched on astrocytic endfeet at blood-brain interfaces. AQP4 is also expressed at cerebrospinal fluid (CSF)-brain interfaces, such as the pial glia limitans and the ependyma and at the choroid plexus blood-CSF barrier. However, little is known regarding pathology at these sites in NMO. Therefore, we evaluated AQP4 expression, microglial reactivity, and complement deposition at pial and ependymal surfaces and in the fourth ventricle choroid plexus in 23 autopsy cases with clinically and/or pathologically confirmed NMO or NMO spectrum disorder. These findings were compared to five cases with multiple sclerosis, five cases of choroid plexus papilloma, and five control cases without central nervous system disease. In the NMO cases, AQP4 immunoreactivity was reduced relative to control levels in the pia (91%; 21/23), ependyma (56%; 9/16), and choroid plexus epithelium (100%; 12/12). AQP4 immunoreactivity was normal in MS cases in these regions. Compared to MS, NMO cases also showed a focal pattern of pial and ependymal complement deposition and more pronounced microglial reactivity. In addition, AQP4 loss, microglial reactivity, and complement deposition colocalized along the pia and ependyma only in NMO cases. Within the choroid plexus, AQP4 loss was coincident with C9neo immunoreactivity on epithelial cell membranes only in NMO cases. These observations demonstrate that NMO immunopathology extends beyond perivascular astrocytic foot processes to include the pia, ependyma, and choroid plexus, suggesting that NMO IgG-induced pathological alterations at CSF-brain and blood-CSF interfaces may contribute to the occurrence of ventriculitis, leptomeningitis, and hydrocephalus observed among NMO patients. Moreover, disruption of the blood-CSF barrier induced by binding of NMO IgG to AQP4 on the basolateral surface of choroid plexus epithelial cells may provide a unique portal for entry of the pathogenic antibody into the central nervous system.


Assuntos
Plexo Corióideo/patologia , Epêndima/patologia , Neuromielite Óptica/patologia , Pia-Máter/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Aquaporina 4/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Líquido Cefalorraquidiano , Plexo Corióideo/metabolismo , Estudos de Coortes , Epêndima/metabolismo , Feminino , Expressão Gênica , Humanos , Masculino , Microglia/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Neuromielite Óptica/metabolismo , Pia-Máter/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Adulto Jovem
12.
Glia ; 64(8): 1331-49, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27189804

RESUMO

Under steady-state conditions the central nervous system (CNS) is traditionally thought to be devoid of antigen presenting cells; however, putative dendritic cells (DCs) expressing enhanced yellow fluorescent protein (eYFP) are present in the retina and brain parenchyma of CD11c-eYFP mice. We previously showed that these mice carry the Crb1(rd8) mutation, which causes retinal dystrophic lesions; therefore we hypothesized that the presence of CD11c-eYFP(+) cells within the CNS may be due to pathology associated with the Crb1(rd8) mutation. We generated CD11c-eYFP Crb1(wt/wt) mice and compared the distribution and immunophenotype of CD11c-eYFP(+) cells in CD11c-eYFP mice with and without the Crb1(rd8) mutation. The number and distribution of CD11c-eYFP(+) cells in the CNS was similar between CD11c-eYFP Crb1(wt/wt) and CD11c-eYFP Crb1(rd8/rd8) mice. CD11c-eYFP(+) cells were distributed throughout the inner retina, and clustered in brain regions that receive input from the external environment or lack a blood-brain barrier. CD11c-eYFP(+) cells within the retina and cerebral cortex of CD11c-eYFP Crb1(wt/wt) mice expressed CD11b, F4/80, CD115 and Iba-1, but not DC or antigen presentation markers, whereas CD11c-eYFP(+) cells within the choroid plexus and pia mater expressed CD11c, I-A/I-E, CD80, CD86, CD103, DEC205, CD8α and CD135. The immunophenotype of CD11c-eYFP(+) cells and microglia within the CNS was similar between CD11c-eYFP Crb1(wt/wt) and CD11c-eYFP Crb1(rd8/rd8) mice; however, CD11c and I-A/I-E expression was significantly increased in CD11c-eYFP Crb1(rd8/rd8) mice. This study demonstrates that the overwhelming majority of CNS CD11c-eYFP(+) cells do not display the phenotype of DCs or their precursors and are most likely a subpopulation of microglia. GLIA 2016. GLIA 2016;64:1331-1349.


Assuntos
Proteínas de Bactérias/metabolismo , Encéfalo/citologia , Antígeno CD11c/metabolismo , Células Dendríticas/citologia , Proteínas Luminescentes/metabolismo , Microglia/citologia , Retina/citologia , Animais , Proteínas de Bactérias/genética , Encéfalo/metabolismo , Células Dendríticas/metabolismo , Citometria de Fluxo , Imunofluorescência , Técnicas Imunoenzimáticas , Antígenos Comuns de Leucócito/metabolismo , Proteínas Luminescentes/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pia-Máter/citologia , Pia-Máter/metabolismo , Retina/metabolismo
13.
Mol Brain ; 8: 53, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26382033

RESUMO

BACKGROUND: Radial glial cells (RGCs), the instructive scaffolds for neuronal migration, are well characterized by their unique morphology and polarization; these cells extend elongated basal processes to the pial basement membrane (BM) and parallel to one another. However, little is known about the mechanisms that underlie the developmental regulation and maintenance of this unique morphology. RESULTS: Here, by crossing Fstl1 (fl/fl) mice with an EIIa-Cre line, we identified a new role for the secreted glycoprotein Follistatin like-1 (FSTL1). The ablation of Fstl1 in both of its cortical expression domains, the ventricular zone (VZ) and the pia mater, resulted in RGC morphologic disruption; basal processes were not parallel to each other, and endfeet exhibited greater density and branching. However, Fstl1 deletion in only the VZ in the Emx1 (IREScre); Fstl1 (fl/fl) line did not affect RGC morphology, indicating that FSTL1 derived from the pia mater might be more important for RGC morphology. In addition, upper-layer projection neurons, not deeper-layer projection neurons, failed to reach their appropriate positions. We also found that BMP, AKT/PKB, Cdc42, GSK3ß, integrin and reelin signals, which have previously been reported to regulate RGC development, were unchanged, indicating that Fstl1 may function through a unique mechanism. CONCLUSIONS: In the present study, we identified a new role for FSTL1 in the development of radial glial scaffolds and the neuronal migration of upper-layer projection neurons. Our findings will improve understanding of the regulation of RGC development and neuronal migration.


Assuntos
Proteínas Relacionadas à Folistatina/metabolismo , Neuroglia/metabolismo , Animais , Membrana Basal/metabolismo , Polaridade Celular , Forma Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/metabolismo , Proteínas Relacionadas à Folistatina/deficiência , Deleção de Genes , Camundongos , Neuroglia/citologia , Pia-Máter/metabolismo , Proteína Reelina
14.
Endocrinology ; 155(5): 2009-19, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24601886

RESUMO

Thyroid hormone regulates immune functions and has antiinflammatory effects. In promoter assays, the thyroid hormone-activating enzyme, type 2 deiodinase (D2), is highly inducible by the inflammatory transcription factor nuclear factor-κ B (NF-κB), but it is unknown whether D2 is induced in a similar fashion in vivo during inflammation. We first reexamined the effect of bacterial lipopolysaccharide (LPS) on D2 expression and NF-κB activation in the rat and mouse brain using in situ hybridization. In rats, LPS induced very robust D2 expression in normally non-D2-expressing cells in the leptomeninges, adjacent brain blood vessels, and the choroid plexus. These cells were vimentin-positive fibroblasts and expressed the NF-κB activation marker, inhibitor κ B-α mRNA, at 2 hours after injection, before the increase in D2 mRNA. In mice, LPS induced intense D2 expression in the choroid plexus but not in leptomeninges, with an early expression peak at 2 hours. Moderate D2 expression along numerous brain blood vessels appeared later. D2 and NF-κB activation was induced in tanycytes in both species but with a different time course. Enzymatic assays from leptomeningeal and choroid plexus samples revealed exceptionally high D2 activity in LPS-treated rats and Syrian hamsters and moderate but significant increases in mice. These data demonstrate the cell type-specific, highly inducible nature of D2 expression by inflammation, and NF-κB as a possible initiating factor, but also warrant attention for species differences. The results suggest that D2-mediated T3 production by fibroblasts regulate local inflammatory actions in the leptomeninges, choroid plexus and brain blood vessels, and perhaps also in other organs.


Assuntos
Plexo Corióideo/metabolismo , Modelos Animais de Doenças , Encefalite/metabolismo , Indução Enzimática , Iodeto Peroxidase/biossíntese , Meninges/metabolismo , Meningite/metabolismo , Animais , Aracnoide-Máter/imunologia , Aracnoide-Máter/metabolismo , Aracnoide-Máter/patologia , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Plexo Corióideo/imunologia , Plexo Corióideo/patologia , Cricetinae , Encefalite/imunologia , Encefalite/patologia , Células Ependimogliais/imunologia , Células Ependimogliais/metabolismo , Células Ependimogliais/patologia , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Masculino , Meninges/imunologia , Meninges/patologia , Meningite/imunologia , Meningite/patologia , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/biossíntese , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Pia-Máter/imunologia , Pia-Máter/metabolismo , Pia-Máter/patologia , Ratos , Ratos Sprague-Dawley , Iodotironina Desiodinase Tipo II
15.
Connect Tissue Res ; 55(2): 147-55, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24409813

RESUMO

Abstract It is very well known that spinal meninges are composed of three layers, dura, arachnoid and pia mater, and that the main components of pia mater are collagen and reticular fibers. However, the distribution of those fibers has not been extensively investigated but just described as a mesh of fibers. In this study, we detected novel structures, which are composed of unidirectionally arranged fibers, in a rat spinal pia mater by using a polarized light microscope. They were seen as three parallel lines, one of which ran along a posterior spinal vein and the rest two of which ran along a pair of posterior spinal arteries. Histological analysis including Masson's trichrome, picrosirius-red staining, Gordon & Sweet's staining and immunohistochemistry with anti-collagen type 1 and 3 antibodies uncovered that they are mainly composed of collagen fibers and some reticular fibers. In addition, a putative primo vessel was detected in the novel fibrous tissue, which was proven out to be different from a blood vessel. In conclusion, we report a newly detected fibrous structure in the spinal pia mater, which may contribute to provide tensile force to the spinal meninges and to harbor the primo vascular system inside.


Assuntos
Colágeno Tipo III/metabolismo , Colágeno Tipo I/metabolismo , Tecido Elástico/metabolismo , Pia-Máter , Medula Espinal , Animais , Masculino , Microscopia de Polarização/métodos , Pia-Máter/irrigação sanguínea , Pia-Máter/citologia , Pia-Máter/metabolismo , Ratos , Ratos Sprague-Dawley , Medula Espinal/irrigação sanguínea , Medula Espinal/citologia , Medula Espinal/metabolismo
17.
Neonatology ; 104(2): 79-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23859876

RESUMO

BACKGROUND: The neurovascular unit encompasses the functional interactions of cerebrovascular and brain parenchymal cells necessary for the metabolic homeostasis of neurons. Previous studies indicated marked but only transient (1-4 h) reactive oxygen species-dependent neurovascular dysfunction in newborn pigs after severe hypoxic/ischemic (H/I) stress contributing to the neuronal injury after birth asphyxia. OBJECTIVES: Our major purpose was to determine if neurovascular dysfunction would also occur later, at 24 h after a milder H/I stress. We also tested if the putative hydroxyl radical scavenger hydrogen (H2) exerted neurovascular protection. METHODS: Anesthetized, ventilated piglets were assigned to three groups of 9 animals: time control, asphyxia/reventilation with air, and asphyxia/reventilation with air +2.1% H2 for 4 h. Asphyxia was induced by suspending ventilation for 8 min. Cerebrovascular reactivity (CR) of pial arterioles was determined using closed cranial window/intravital microscopy 24 h after asphyxia to the endothelium-dependent cerebrovascular stimulus hypercapnia, the neuronal function-dependent stimulus N-methyl-D-aspartate (NMDA), norepinephrine, and sodium nitroprusside. The brains were subjected to histopathology. RESULTS: Hemodynamic parameters, blood gases, and core temperature did not differ significantly among the experimental groups. In the early reventilation period, the recovery of electroencephalographic activity was significantly better in H2-treated animals. Asphyxia/reventilation severely attenuated CR to hypercapnia and NMDA; however, reactivity to norepinephrine and sodium nitroprusside were unaltered. H2 fully or partially preserved CR to hypercapnia or NMDA, respectively. Histopathology revealed modest neuroprotection afforded by H2. CONCLUSIONS: Severe stimulus-selective delayed neurovascular dysfunction develops and persists even after mild H/I stress. H2 alleviates this delayed neurovascular dysfunction that can contribute to its neuroprotective effect.


Assuntos
Asfixia Neonatal/tratamento farmacológico , Sequestradores de Radicais Livres/farmacologia , Hidrogênio/farmacologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Pia-Máter/irrigação sanguínea , Pia-Máter/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Arteríolas/efeitos dos fármacos , Arteríolas/fisiopatologia , Asfixia Neonatal/metabolismo , Asfixia Neonatal/patologia , Asfixia Neonatal/fisiopatologia , Gasometria , Ondas Encefálicas/efeitos dos fármacos , Modelos Animais de Doenças , Eletroencefalografia , Agonistas de Aminoácidos Excitatórios/farmacologia , Hemodinâmica/efeitos dos fármacos , Radical Hidroxila/metabolismo , Hipercapnia/fisiopatologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Pia-Máter/metabolismo , Pia-Máter/patologia , Recuperação de Função Fisiológica , Respiração Artificial , Suínos , Fatores de Tempo , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
18.
J Vasc Res ; 50(4): 332-45, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23860357

RESUMO

OBJECTIVE: The aim of this study was to assess the in vivo structural and functional remodeling of pial arteriolar networks in the ischemic area of rats submitted to transient middle cerebral artery occlusion (MCAO) and different time intervals of reperfusion. METHODS AND RESULTS: Two closed cranial windows were implanted above the left and right parietal cortex to observe pial microcirculation by fluorescence microscopy. The geometric characteristics of pial arteriolar networks, permeability increase, leukocyte adhesion and capillary density were analyzed after 1 h or 1, 7, 14 or 28 days of reperfusion. MCAO and 1-hour reperfusion caused marked microvascular changes in pial networks. The necrotic core was devoid of vessels, while the penumbra area presented a few arterioles, capillaries and venules with severe neuronal damage. Penumbra microvascular permeability and leukocyte adhesion were pronounced. At 7 days of reperfusion, new pial arterioles were organized in anastomotic vessels, overlapping the ischemic core and in penetrating pial arterioles. Vascular remodeling caused different arteriolar rearrangement up to 28 days of reperfusion and animals gradually regained their motor and sensory functions. CONCLUSIONS: Transient MCAO-induced pial-network remodeling is characterized by arteriolar anastomotic arcades. Remodeling mechanisms appear to be accompanied by an increased expression of nitric oxide synthases.


Assuntos
Capilares/fisiopatologia , Circulação Cerebrovascular , Infarto da Artéria Cerebral Média/terapia , Microcirculação , Pia-Máter/irrigação sanguínea , Reperfusão , Animais , Arteríolas/fisiopatologia , Comportamento Animal , Capilares/patologia , Permeabilidade Capilar , Adesão Celular , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Leucócitos/patologia , Masculino , Microscopia de Fluorescência , Microscopia de Vídeo , Atividade Motora , Necrose , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Pia-Máter/metabolismo , Pia-Máter/patologia , Ratos , Ratos Wistar , Recuperação de Função Fisiológica , Sensação , Índice de Gravidade de Doença , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
J Anat ; 220(5): 435-46, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22352427

RESUMO

Podoplanin is a transmembrane glycoprotein indirectly linked to classic cadherins through ezrin-actin networks. Recently, the overexpression of podoplanin in high-grade malignancy brain tumors has been reported. The aim of this study was to investigate the expression of podoplanin and classic cadherins in the mouse brain. Immunohistochemistry showed that podoplanin was expressed on ependymal cells and choroid plexus epithelial cells at the ventricle side of the cell surface and at the cell-cell junctions, and on retinal pigment epithelial cells and in the pia mater; P-cadherin between choroid plexus epithelial cells and endothelial cells at the basement membrane side of cell surface, and between retinal pigment epithelial cells; VE-cadherin on the PECAM-1 positive-choroid plexus endothelial cells of the fibrovascular core; and N-cadherin on the cell surface and at the cell-cell junctions of ependymal cells, and in the pia mater. The regions expressing podoplanin, P-cadherin, and VE-cadherin did not coincide. In real-time PCR analysis, the amounts of podoplanin and P- and N-cadherin mRNA were larger in the ventricular wall with choroid plexus than in the abdominal aorta and cerebrum. In the RT-PCR analysis, the intensities of amplicon for VE-cadherin mRNA were the same for the abdominal aorta, cerebrum, and ventricular wall with the choroid plexus, suggesting that mouse ependymal cells, choroid plexus epithelial cells, and glial cells under the pia mater have the ability to express podoplanin and P- and N-cadherins. Glial cells and retinal pigment epithelial cells may create barriers by podoplanin and classic cadherins as a rate-determining step for transmission of blood components.


Assuntos
Caderinas/metabolismo , Plexo Corióideo/metabolismo , Glicoproteínas de Membrana/metabolismo , Pia-Máter/metabolismo , Animais , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Reação em Cadeia da Polimerase , Epitélio Pigmentado da Retina/metabolismo
20.
Stem Cells Dev ; 21(13): 2350-4, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22339778

RESUMO

Increasing evidence indicates that neural stem/progenitor cells (NSPCs) reside in many regions of the central nervous system (CNS), including the subventricular zone (SVZ) of the lateral ventricle, subgranular zone of the hippocampal dentate gyrus, cortex, striatum, and spinal cord. Using a murine model of cortical infarction, we recently demonstrated that the leptomeninges (pia mater), which cover the entire cortex, also exhibit NSPC activity in response to ischemia. Pial-ischemia-induced NSPCs expressed NSPC markers such as nestin, formed neurosphere-like cell clusters with self-renewal activity, and differentiated into neurons, astrocytes, and oligodendrocytes, although they were not identical to previously reported NSPCs, such as SVZ astrocytes, ependymal cells, oligodendrocyte precursor cells, and reactive astrocytes. In this study, we showed that leptomeningeal cells in the poststroke brain express the immature neuronal marker doublecortin as well as nestin. We also showed that these cells can migrate into the poststroke cortex. Thus, the leptomeninges may participate in CNS repair in response to brain injury.


Assuntos
Encéfalo/patologia , Proteínas de Filamentos Intermediários/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Pia-Máter/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Encéfalo/metabolismo , Movimento Celular , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Proteínas do Domínio Duplacortina , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/genética , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas do Tecido Nervoso/genética , Nestina , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurogênese , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeos/genética , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Pia-Máter/patologia , Acidente Vascular Cerebral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA