Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 194
Filtrar
1.
Stroke ; 53(3): 987-998, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35144488

RESUMO

BACKGROUND: Promotion of hematoma resolution in a timely manner reduces intracerebral hemorrhage (ICH) brain injury induced by toxic blood components and subsequent neuroinflammation. The meningeal lymphatic system is responsible for clearance of macromolecules and pathogenic substances from the central nervous system; however, its role in intraparenchymal hematoma clearance and ICH outcomes is unknown. In the present study, we aimed to understand the contribution of the meningeal lymphatic system to ICH pathologies and to test whether pharmacological enhancement of meningeal lymphatic function promotes hematoma resolution and brain recovery after ICH. METHODS: Immunofluorescence of whole-mount meninges was used to measure complexity and coverage level of meningeal lymphatic vasculature following ICH induction. Fluorescent microbeads and PKH-26-labeled erythrocytes were used to evaluate drainage function of the meningeal lymphatic system. Visudyne treatment, deep cervical lymph node ligation, and VEGF (vascular endothelial growth factor)-C injection were performed to manipulate meningeal lymphatic function. Neurobehavioral performance and hematoma volume were assayed by the cylinder test and histological measurements. Iron deposition, residual erythrocytes, neuronal loss, and astrogliosis were assessed by immunohistochemistry and antibody-based fluorescence staining. RESULTS: Meningeal lymphangiogenesis and enhanced lymphatic drainage occurred during the late phase of ICH. Ablation and blockage of meningeal lymphatic vessels impeded hematoma clearance, whereas pharmacological enhancement of their function reduced hematoma volume, improved behavioral performance, and reduced brain residual erythrocytes, iron deposition, neuronal loss, and astroglial activation. CONCLUSIONS: Early enhancement of meningeal lymphatic function is beneficial for ICH recovery. Targeting the meningeal lymphatic system is therefore a potential therapeutic approach for treating ICH.


Assuntos
Encéfalo/patologia , Hemorragia Cerebral/patologia , Linfangiogênese/fisiologia , Sistema Linfático/patologia , Meninges/patologia , Animais , Encéfalo/efeitos dos fármacos , Hemorragia Cerebral/tratamento farmacológico , Cilostazol/farmacologia , Cilostazol/uso terapêutico , Linfangiogênese/efeitos dos fármacos , Sistema Linfático/efeitos dos fármacos , Masculino , Meninges/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
2.
Artigo em Inglês | MEDLINE | ID: mdl-34911793

RESUMO

BACKGROUND AND OBJECTIVES: To investigate whether the formation or retention of meningeal ectopic lymphoid tissue (mELT) can be inhibited by the sphingosine 1-phosphate receptor 1,5 modulator siponimod (BAF312) in a murine model of multiple sclerosis (MS). METHODS: A murine spontaneous chronic experimental autoimmune encephalomyelitis (EAE) model, featuring meningeal inflammatory infiltrates resembling those in MS, was used. To prevent or treat EAE, siponimod was administered daily starting either before EAE onset or at peak of disease. The extent and cellular composition of mELT, the spinal cord parenchyma, and the spleen was assessed by histology and immunohistochemistry. RESULTS: Siponimod, when applied before disease onset, ameliorated EAE. This effect was also present, although less prominent, when treatment started at peak of disease. Treatment with siponimod resulted in a strong reduction of the extent of mELT in both treatment paradigms. Both B and T cells were diminished in the meningeal compartment. DISCUSSION: Beneficial effects on the disease course correlated with a reduction in mELT, suggesting that inhibition of mELT may be an additional mechanism of action of siponimod in the treatment of EAE. Further studies are needed to establish causality and confirm this observation in MS.


Assuntos
Azetidinas/farmacologia , Compostos de Benzil/farmacologia , Encefalomielite Autoimune Experimental , Meninges/efeitos dos fármacos , Esclerose Múltipla , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacologia , Estruturas Linfoides Terciárias , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/prevenção & controle , Humanos , Meninges/imunologia , Camundongos , Esclerose Múltipla/complicações , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/prevenção & controle , Estruturas Linfoides Terciárias/tratamento farmacológico , Estruturas Linfoides Terciárias/etiologia , Estruturas Linfoides Terciárias/prevenção & controle
3.
Int J Mol Sci ; 22(19)2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34638999

RESUMO

Neural precursors (NPs) present in the hippocampus can be modulated by several neurogenic stimuli, including environmental enrichment (EE) acting through BDNF-TrkB signaling. We have recently identified NPs in meninges; however, the meningeal niche response to pro-neurogenic stimuli has never been investigated. To this aim, we analyzed the effects of EE exposure on NP distribution in mouse brain meninges. Following neurogenic stimuli, although we did not detect modification of the meningeal cell number and proliferation, we observed an increased number of neural precursors in the meninges. A lineage tracing experiment suggested that EE-induced ß3-Tubulin+ immature neuronal cells present in the meninges originated, at least in part, from GLAST+ radial glia cells. To investigate the molecular mechanism responsible for meningeal reaction to EE exposure, we studied the BDNF-TrkB interaction. Treatment with ANA-12, a TrkB non-competitive inhibitor, abolished the EE-induced meningeal niche changes. Overall, these data showed, for the first time, that EE exposure induced meningeal niche remodeling through TrkB-mediated signaling. Fluoxetine treatment further confirmed the meningeal niche response, suggesting it may also respond to other pharmacological neurogenic stimuli. A better understanding of the neurogenic stimuli modulation for meninges may be useful to improve the effectiveness of neurodegenerative and neuropsychiatric treatments.


Assuntos
Microambiente Celular , Meio Ambiente , Glicoproteínas de Membrana/metabolismo , Meninges/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Imunofluorescência , Fluoxetina/farmacologia , Meninges/efeitos dos fármacos , Meninges/patologia , Camundongos , Neuroglia/metabolismo , Neurônios/metabolismo
4.
Front Immunol ; 12: 688254, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093593

RESUMO

Several barriers separate the central nervous system (CNS) from the rest of the body. These barriers are essential for regulating the movement of fluid, ions, molecules, and immune cells into and out of the brain parenchyma. Each CNS barrier is unique and highly dynamic. Endothelial cells, epithelial cells, pericytes, astrocytes, and other cellular constituents each have intricate functions that are essential to sustain the brain's health. Along with damaging neurons, a traumatic brain injury (TBI) also directly insults the CNS barrier-forming cells. Disruption to the barriers first occurs by physical damage to the cells, called the primary injury. Subsequently, during the secondary injury cascade, a further array of molecular and biochemical changes occurs at the barriers. These changes are focused on rebuilding and remodeling, as well as movement of immune cells and waste into and out of the brain. Secondary injury cascades further damage the CNS barriers. Inflammation is central to healthy remodeling of CNS barriers. However, inflammation, as a secondary pathology, also plays a role in the chronic disruption of the barriers' functions after TBI. The goal of this paper is to review the different barriers of the brain, including (1) the blood-brain barrier, (2) the blood-cerebrospinal fluid barrier, (3) the meningeal barrier, (4) the blood-retina barrier, and (5) the brain-lesion border. We then detail the changes at these barriers due to both primary and secondary injury following TBI and indicate areas open for future research and discoveries. Finally, we describe the unique function of the pro-inflammatory cytokine interleukin-1 as a central actor in the inflammatory regulation of CNS barrier function and dysfunction after a TBI.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematorretiniana/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Interleucina-1/metabolismo , Meninges/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/imunologia , Barreira Hematorretiniana/patologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/patologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/antagonistas & inibidores , Interleucina-1/antagonistas & inibidores , Meninges/efeitos dos fármacos , Meninges/imunologia , Meninges/patologia , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais
5.
Artigo em Inglês | MEDLINE | ID: mdl-34021057

RESUMO

OBJECTIVE: To investigate whether anti-CD20 B-cell-depleting monoclonal antibodies (ɑCD20 mAbs) inhibit the formation or retention of meningeal ectopic lymphoid tissue (mELT) in a murine model of multiple sclerosis (MS). METHODS: We used a spontaneous chronic experimental autoimmune encephalomyelitis (EAE) model of mice with mutant T-cell and B-cell receptors specific for myelin oligodendrocyte glycoprotein (MOG), which develop meningeal inflammatory infiltrates resembling those described in MS. ɑCD20 mAbs were administered in either a preventive or a treatment regimen. The extent and cellular composition of mELT was assessed by histology and immunohistochemistry. RESULTS: ɑCD20 mAb, applied in a paradigm to either prevent or treat EAE, did not alter the disease course in either condition. However, ɑCD20 mAb depleted virtually all B cells from the meningeal compartment but failed to prevent the formation of mELT altogether. Because of the absence of B cells, mELT was less densely populated with immune cells and the cellular composition was changed, with increased neutrophil granulocytes. CONCLUSIONS: These results demonstrate that, in CNS autoimmune disease, meningeal inflammatory infiltrates may form and persist in the absence of B cells. Together with the finding that ɑCD20 mAb does not ameliorate spontaneous chronic EAE with mELT, our data suggest that mELT may have yet unknown capacities that are independent of B cells and contribute to CNS autoimmunity.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD20/imunologia , Linfócitos B , Encefalomielite Autoimune Experimental , Fatores Imunológicos/farmacologia , Meninges , Estruturas Linfoides Terciárias , Animais , Anticorpos Monoclonais/administração & dosagem , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Fatores Imunológicos/administração & dosagem , Meninges/efeitos dos fármacos , Meninges/imunologia , Camundongos , Camundongos Transgênicos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Esclerose Múltipla/prevenção & controle , Glicoproteína Mielina-Oligodendrócito , Estruturas Linfoides Terciárias/tratamento farmacológico , Estruturas Linfoides Terciárias/imunologia
6.
Commun Biol ; 4(1): 260, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637884

RESUMO

Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.


Assuntos
Anti-Inflamatórios/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Plexo Corióideo/efeitos dos fármacos , Digoxina/farmacologia , Células Endoteliais/efeitos dos fármacos , Inflamação/tratamento farmacológico , Lanatosídeos/farmacologia , Meninges/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Células Cultivadas , Plexo Corióideo/metabolismo , Plexo Corióideo/patologia , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Ensaios de Triagem em Larga Escala , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Meninges/metabolismo , Meninges/patologia , Pericitos/metabolismo , Pericitos/patologia , Técnicas de Cultura de Tecidos
7.
Biomed Res Int ; 2020: 4616308, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33015166

RESUMO

BACKGROUND: The brain is in many ways an immunologically and pharmacologically privileged site because of the blood-brain barrier (BBB). But for chronic peripheral inflammation, inflammatory signals can be transmitted from the peripheral system into the central nervous system (CNS) through multiple channels and result in neuroinflammation. Leptomeningeal cells that form the BBB can trigger one signaling pathway by releasing cytokines to transmit inflammatory signals. Besides, the Janus kinase (JAK) family may have a certain function in the activation of leptomeninges. In the present study, we try to use coniferyl aldehyde (CA), a natural anti-inflammatory phenolic compound, to inhibit this inflammatory process and elucidate the underlying molecular mechanisms. RESULTS: Secretion of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6) significantly increased after incubation with P. gingivalis. Moreover, TNF-α, IL-1ß, and IL-6 levels were upregulated, and the JAK2 signaling was enhanced in leptomeningeal cells in a conditioned medium from activated macrophages, which leads to the immune response in microglia. However, this inflammatory effect of leptomeningeal cells was reversed by CA administration, accompanied by the decreased immune response in microglia. The western blot assay revealed that JAK2 phosphorylation was suppressed in leptomeningeal cells treated with CA. CONCLUSIONS: This study demonstrates that activated macrophages by P. gingivalis markedly induce the release of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6) from leptomeningeal cells, thereby activating the JAK2 signaling pathway and subsequently enhancing immune responses in microglia in the CNS. CA effectively inhibits the inflammatory effect of leptomeningeal cells via suppressing the JAK2 signaling pathway.


Assuntos
Acroleína/análogos & derivados , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Janus Quinase 2/metabolismo , Meninges/efeitos dos fármacos , Acroleína/farmacologia , Animais , Linhagem Celular , Citocinas/metabolismo , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Meninges/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
8.
Tuberculosis (Edinb) ; 122: 101924, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32501258

RESUMO

Tuberculous meningitis (TBM) is the most devastating form of TB, resulting in death or neurological disability in up to 50% of patients affected. Treatment is similar to that of pulmonary TB, despite poor cerebrospinal fluid (CSF) penetration of the cornerstone anti-TB drug rifampicin. Considering TBM pathology, it is critical that optimal drug concentrations are reached in the meninges, brain and/or the surrounding CSF. These type of data are difficult to collect in TBM patients. This review aims to identify and describe a preclinical model representative for human TBM which can provide the indispensable data needed for future pharmacological characterization and prioritization of new TBM regimens in the clinical setting. We reviewed existing literature on treatment of TBM in preclinical models: only eight articles, all animal studies, could be identified. None of the animal models completely recapitulated human disease and in most of the animal studies key pharmacokinetic data were missing, making the comparison with human exposure and CNS distribution, and the study of pharmacokinetic-pharmacodynamic relationships impossible. Another 18 articles were identified using other bacteria to induce meningitis with treatment including anti-TB drugs (predominantly rifampicin, moxifloxacin and levofloxacin). Of these articles the pharmacokinetics, i.e. plasma exposure and CSF:plasma ratios, of TB drugs in meningitis could be evaluated. Exposures (except for levofloxacin) agreed with human exposures and also most CSF:plasma ratios agreed with ratios in humans. Considering the lack of an ideal preclinical pharmacological TBM model, we suggest a combination of 1. basic physicochemical drug data combined with 2. in vitro pharmacokinetic and efficacy data, 3. an animal model with adequate pharmacokinetic sampling, microdialysis or imaging of drug distribution, all as a base for 4. physiologically based pharmacokinetic (PBPK) modelling to predict response to TB drugs in treatment of TBM.


Assuntos
Antituberculosos/farmacologia , Meninges/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Tuberculose Meníngea/tratamento farmacológico , Animais , Antituberculosos/farmacocinética , Antituberculosos/toxicidade , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Humanos , Meninges/microbiologia , Mycobacterium tuberculosis/patogenicidade , Especificidade da Espécie , Tuberculose Meníngea/microbiologia
9.
Cephalalgia ; 39(11): 1358-1365, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31475573

RESUMO

BACKGROUND: Botulinum neurotoxin type A, an FDA-approved prophylactic drug for chronic migraine, is thought to achieve its therapeutic effect through blocking activation of unmyelinated meningeal nociceptors and their downstream communications with myelinated nociceptors and potentially the vasculature and immune cells. Prior investigations to determine botulinum neurotoxin type A effects on meningeal nociceptors were carried out in male rats and tested with stimuli that act outside the blood brain barrier. Here, we sought to explore the effects of extracranial injections of botulinum neurotoxin type A on activation of meningeal nociceptors by cortical spreading depression, an event which occurs inside the blood brain barrier, in female rats. MATERIAL AND METHODS: Using single-unit recording, we studied myelinated C- and unmyelinated Aδ-meningeal nociceptors' responses to cortical spreading depression 7-14 days after injection of botulinum neurotoxin type A or saline along calvarial sutures. RESULTS: In female rats, responses to cortical spreading depression were typically more prolonged and, in some cases, began at relatively longer latencies post-cortical spreading depression, than had been observed in previous studies in male rats. Extracranial administration of botulinum neurotoxin type A reduced significantly the prolonged firing of the meningeal nociceptors, in the combined sample of Aδ- and C-fiber, but not their response probability. DISCUSSION: The findings suggest that the mechanism of action by which botulinum neurotoxin type A prevents migraine differ from the one by which calcitonin gene-related peptide monoclonal antibodies prevent migraine and that even when the origin of migraine is central (i.e. in the cortex), a peripherally acting drug can intercept/prevent the headache.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Meninges/efeitos dos fármacos , Fármacos Neuromusculares/farmacologia , Nociceptores/efeitos dos fármacos , Animais , Feminino , Fibras Nervosas Amielínicas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
10.
Mater Sci Eng C Mater Biol Appl ; 102: 34-44, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31147006

RESUMO

Spinal cord injury (SCI) is one of the most precarious conditions which have been one of the major reasons for continuous increasing mortality rate of SCI patients. Currently, there is no effective treatment modality for SCI patients posing major threat to the scientific and medical community. The available strategies don't mimic with the natural processes of nervous tissues repair/regeneration and majority of the approaches may induce the additional fibrotic or immunological response at the injury site and are not readily available on demand. To overcome these hurdles, we have developed a ready to use bioengineered human functional neurological construct (BHNC) for regenerative applications in SCI defects. We used cryopreserved meningeal tissues (CMT) for bioengineering these neurological constructs using acellularization and repopulation technology. The technology adopted herein generates intact neurological scaffolds from CMT and retains several crucial structural, biochemical and mechanical cues to enhance the regenerative mechanisms. The neurogenic differentiation on CMT scaffolds was almost similar to the freshly prepared meningeal scaffolds and mimics with the natural nervous tissue developmental mechanisms which offer intact 3D-microarchitecture and hospitable microenvironment enriched with several crucial neurotrophins for long-term cell survival and function. Functional assessment of developed BHNC showed highly increased positive staining for pre-synaptic granules of Synapsis-1 along with MAP-2 antibody with punctuate distribution in axonal regions of the neuronal cells which was well supported by the gene expression analysis of functional transcripts. Given the significant improvement in the field may enable to generate more such ready to use functional BHNC for wider applicability in SCI repair/regeneration.


Assuntos
Materiais Biomiméticos/farmacologia , Criopreservação , Meninges/fisiologia , Engenharia Tecidual/métodos , Tecidos Suporte/química , Fenômenos Biomecânicos , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Humanos , Meninges/efeitos dos fármacos , Meninges/ultraestrutura , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Neuropharmacology ; 149: 113-123, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30768945

RESUMO

BACKGROUND: Recent discovery of mechanosensitive Piezo receptors in trigeminal ganglia suggested the novel molecular candidate for generation of migraine pain. However, the contribution of Piezo channels in migraine pathology was not tested yet. Therefore, in this study, we explored a potential involvement of Piezo channels in peripheral trigeminal nociception implicated in generation of migraine pain. METHODS: We used immunohistochemistry, calcium imaging, calcitonin gene related peptide (CGRP) release assay and electrophysiology in mouse and rat isolated trigeminal neurons and rat hemiskulls to study action of various stimulants of Piezo receptors on migraine-related peripheral nociception. RESULTS: We found that essential (35%) fraction of isolated rat trigeminal neurons responded to chemical Piezo1 agonist Yoda1 and about a half of Yoda1 positive neurons responded to hypo-osmotic solution (HOS) and a quarter to mechanical stimulation by focused ultrasound (US). In ex vivo hemiskull preparation, Yoda1 and HOS largely activated persistent nociceptive firing in meningeal branches of trigeminal nerve. By using our novel cluster analysis of pain spikes, we demonstrated that 42% of fibers responded to Piezo1 agonist and 20% of trigeminal fibers were activated by Yoda1 and by capsaicin, suggesting expression of Piezo receptors in TRPV1 positive peptidergic nociceptive nerve fibers. Consistent with this, Yoda1 promoted the release of the key migraine mediator CGRP from hemiskull preparation. CONCLUSION: Taken together, our data suggest the involvement of mechanosensitive Piezo receptors, in particular, Piezo1 subtype in peripheral trigeminal nociception, which provides a new view on mechanotransduction in migraine pathology and suggests novel molecular targets for anti-migraine medicine.


Assuntos
Canais Iônicos/agonistas , Meninges/fisiologia , Transtornos de Enxaqueca/metabolismo , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Animais , Cálcio , Canais Iônicos/metabolismo , Mecanorreceptores/fisiologia , Mecanotransdução Celular , Meninges/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Aferentes/metabolismo , Nociceptores/fisiologia , Dor , Cultura Primária de Células , Ratos , Ratos Wistar , Células Receptoras Sensoriais , Nervo Trigêmeo/fisiologia
12.
AJNR Am J Neuroradiol ; 39(9): 1739-1744, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30049717

RESUMO

BACKGROUND AND PURPOSE: Propofol is a cerebral vasoconstrictor that modulates cerebral perfusion by decreasing the metabolic rate of oxygen. Because younger children often undergo intravenous sedation for MR imaging, this study set out to evaluate the degree of leptomeningeal contrast enhancement on 3T postcontrast brain MR imaging and to determine whether this phenomenon relates to sequence, sedation dosage, or patient age or weight. MATERIALS AND METHODS: During a 2-year period, of 152 children 1-5 years of age who underwent MR imaging, 43 were included for MRI review. Of these, 37 underwent postcontrast imaging with either solely gradient-echo T1WI (n = 20) or spin-echo T1WI (n = 17); notably, 6 patients underwent both sequences. Three neuroradiologists separately graded the degree of leptomeningeal contrast enhancement (grades 0-3) that was correlated with various factors and calculated the interobserver reliability. RESULTS: For the 43 patients, the mean patient age was 3.1 ± 1.4 years. The leptomeningeal contrast-enhancement grade was significantly greater (P < .0001) on spin-echo T1WI (1.9-2.1) versus gradient-echo TIWI (1.2-1.4). Patient weight (r = -0.366 to -.418, P = .003-.01) and age (r = -0.315 to -0.418, P = .004-.032) moderately and inversely correlated with the leptomeningeal contrast-enhancement grade, while the propofol dosage, sedation duration, and time to T1WI post-contrast administration did not (each, P > .05). The interobserver κ was strong regarding the leptomeningeal contrast-enhancement grade on both spin-echo T1WI (κ = 0.609-0.693, P < .0001) and gradient-echo TIWI (κ = 0.567-0.698, P < .0001). CONCLUSIONS: Leptomeningeal contrast enhancement (or "pseudo"-leptomeningeal contrast enhancement) occurs with a greater frequency and degree on 3T postcontrast spin-echo T1WI relative to gradient-echo TIWI in younger children sedated with propofol and should not be mistaken for disease. This phenomenon may be more prominent with lower age or size and may arise from propofol-induced vascular smooth-muscle dilation.


Assuntos
Artefatos , Hipnóticos e Sedativos/efeitos adversos , Imageamento por Ressonância Magnética , Meninges/efeitos dos fármacos , Propofol/efeitos adversos , Criança , Pré-Escolar , Meios de Contraste , Feminino , Humanos , Lactente , Imageamento por Ressonância Magnética/métodos , Masculino , Meninges/diagnóstico por imagem , Neuroimagem/métodos , Reprodutibilidade dos Testes
13.
Oncogene ; 37(36): 4955-4963, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29789719

RESUMO

Somatic activating mutations of smoothened (SMO), a component of the embryonic sonic hedgehog (SHH) signaling pathway, are found in 3-5% of grade I meningiomas, most of them corresponding to meningothelial meningiomas located at the anterior skull base. By generating different developmental stage-specific conditional activations in mice, we define a restricted developmental window during which conditional activation of Smo in Prostaglandin D2-synthase-positive mesoderm-derived meningeal layer of the skull base results in meningothelial meningioma formation. We show a selective vulnerability of the arachnoid from the skull base to Smo activation to initiate tumor development. This prenatal period and specific topography are correlated to the timing and location of SHH signaling involvement in the formation of craniofacial and meninges patterning, strongly corroborating the hypothesis of a developmental origin for Smo-activated meningiomas. Finally, we provide preclinical in vitro evidence of the efficacy of the SMO-inhibitor Sonidegib, supporting further preclinical and clinical evaluation of targeted treatment for refractory SMO-mutant meningiomas.


Assuntos
Neoplasias Meníngeas/metabolismo , Meninges/metabolismo , Meningioma/metabolismo , Base do Crânio/metabolismo , Receptor Smoothened/metabolismo , Animais , Compostos de Bifenilo/farmacologia , Linhagem Celular , Proteínas Hedgehog/metabolismo , Neoplasias Meníngeas/tratamento farmacológico , Meninges/efeitos dos fármacos , Meningioma/tratamento farmacológico , Camundongos , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Base do Crânio/efeitos dos fármacos
14.
Neuropeptides ; 69: 46-52, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29661478

RESUMO

Adriamycin is a potent anthracycline-type antitumor agent, but it also exerts potentially serious side effects due to its cardiotoxic and neurotoxic propensity. Multiple impairments in sensory nerve functions have been recently reported in various rat models. The present experiments were initiated in an attempt to reveal adriamycin-induced changes in sensory effector functions of chemosensitive meningeal afferents. Meningeal blood flow was measured with laser Doppler flowmetry in the parietal dura mater of adult male Wistar rats. The dura mater was repeatedly stimulated by topical applications of capsaicin, a transient receptor potential vanilloid 1 (TRPV1) receptor agonist, or acrolein, a transient receptor potential ankyrin 1 (TRPA1) receptor agonist, which induce the release of calcitonin gene-related peptide (CGRP) from meningeal afferents. The blood flow increasing effects of CGRP, histamine, acetylcholine and forskolin were also measured. Capsaicin- and acrolein-induced CGRP release was measured with enzyme-linked immunoassay in an ex vivo dura mater preparation. TRPV1 content of trigeminal ganglia and TRPV1-, CGRP- and CGRP receptor component-immunoreactive structures were examined in dura mater samples obtained from control and adriamycin-treated rats. The vasodilator effects of capsaicin, acrolein and CGRP were significantly reduced in adriamycin-treated animals while histamine-, acetylcholine- and forskolin-induced vasodilatation were unaffected. Measurements of CGRP release in an ex vivo dura mater preparation revealed an altered dynamic upon repeated stimulations of TRPV1 and TRPA1 receptors. In whole-mount dura mater preparations immunohistochemistry revealed altered CGRP receptor component protein (RCP)-immunoreactivity in adriamycin-treated animals, while CGRP receptor activity modifying protein (RAMP1)-, TRPV1- and CGRP-immunostaining were left apparently unaltered. Adriamycin-treatment slightly reduced TRPV1 protein content of trigeminal ganglia. The present findings demonstrate that adriamycin-treatment alters the function of the trigeminovascular system leading to reduced meningeal sensory neurogenic vasodilatation that may affect the local regulatory and protective mechanisms of chemosensitive afferents leading to alterations in tissue integrity.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Doxorrubicina/toxicidade , Meninges/efeitos dos fármacos , Meninges/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Acroleína/administração & dosagem , Animais , Capsaicina/administração & dosagem , Masculino , Meninges/irrigação sanguínea , Neurônios Aferentes/metabolismo , Ratos Wistar , Canal de Cátion TRPA1/agonistas , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/metabolismo , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo
15.
J Stroke Cerebrovasc Dis ; 27(5): 1237-1251, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29337049

RESUMO

BACKGROUND: Development of collateral circulation after acute ischemic stroke is triggered by shear stress that occurs in pre-existing arterioles. Recently, sphingosine-1-phosphate receptor 1 (S1P1) on endothelial cells was reported to sense shear stress and transduce its signaling pathways. METHODS: BALB/c mice (n = 118) were subjected to permanent middle cerebral artery occlusion (pMCAO) or sham operation. We investigated the effect of an S1P1-selective agonist SEW2871 on leptomeningeal collateral arteries and neurological outcome after pMCAO. RESULTS: Immunohistochemistry showed that without treatment, the expression of S1P1 on endothelial cells of leptomeningeal arteries and capillaries increased early after pMCAO, peaking at 6 hours, whereas a significant increase in the expression of S1P1 in neurons was seen from 24 hours later. After intraperitoneal administration of SEW2871 for 7 days after pMCAO, the number of leptomeningeal collateral arteries was significantly increased, cerebral blood flow improved, infarct volume was decreased, and neurological outcome improved compared with the controls. Significantly increased phosphorylation of endothelial nitric oxide synthase (eNOS) as early as 6 hours after pMCAO and higher expression of tight junction proteins at postoperative day 3 were observed with SEW2871 treatment as assessed by Western blot. Daily administration of SEW2871 also increased capillary density in peri-infarct regions and promoted monocyte/macrophage mobilization to the surface of ischemic cortex at 7 days after pMCAO. CONCLUSIONS: An S1P1-selective agonist enhanced leptomeningeal collateral circulation via eNOS phosphorylation and promoted postischemic angiogenesis with reinforced blood-brain barrier integrity in a mouse model of acute ischemic stroke, leading to smaller infarct volume and better neurological outcome.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Circulação Colateral/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Meninges/irrigação sanguínea , Meninges/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Tiofenos/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Linhagem Celular , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Meninges/metabolismo , Meninges/patologia , Camundongos Endogâmicos BALB C , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Receptores de Lisoesfingolipídeo/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato , Proteínas de Junções Íntimas/metabolismo , Fatores de Tempo
16.
Neuroreport ; 29(1): 19-24, 2018 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-29194293

RESUMO

Emotional and cognitive deficits and associated hippocampal damage observed in multiple sclerosis (MS) are now recognized as primary disease manifestations. However, the pathological substrate of these dysfunctions is unclear. In the experimental autoimmune encephalomyelitis (EAE) MS model, impaired hippocampal-dependent functions are concomitant with severe microglial reactivity and neurodegeneration, but reports vary with respect to evidence of lymphocytic infiltration, raising questions as to the nature of the underlying neurodegenerative mechanisms. Our investigations of EAE-induced inflammation across the hippocampal formation showed CD3 infiltration only in regions adjacent to inflamed meningeal membranes interposed between the ventral aspect of the hippocampus and the dorsal aspect of the mid-brain, but widespread microglial reactivity across the structure. Regions that contact the lateral ventricles do not show inflammation, but CD3 cells are observed in the adjacent ventricular space and choroid plexus, suggesting that microglial reactivity in these regions results from exposure to proinflammatory mediators released into the ventricles. These data indicate that multiple pathophysiological mechanisms underlie hippocampal damage during EAE. Treatment with the immunomodulator FTY720 eliminates microglial reactivity across the whole structure, suggesting potential benefit for neuropsychological symptoms in MS.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Hipocampo/imunologia , Hipocampo/patologia , Animais , Complexo CD3/metabolismo , Plexo Corióideo/efeitos dos fármacos , Plexo Corióideo/imunologia , Plexo Corióideo/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Feminino , Cloridrato de Fingolimode/farmacologia , Hipocampo/efeitos dos fármacos , Imuno-Histoquímica , Imunossupressores/farmacologia , Ventrículos Laterais/efeitos dos fármacos , Ventrículos Laterais/imunologia , Ventrículos Laterais/patologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/patologia , Meninges/efeitos dos fármacos , Meninges/imunologia , Meninges/patologia , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/patologia , Cadeia B de alfa-Cristalina/metabolismo
17.
J Exp Med ; 214(12): 3645-3667, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29141865

RESUMO

The recent discovery of meningeal lymphatic vessels (LVs) has raised interest in their possible involvement in neuropathological processes, yet little is known about their development or maintenance. We show here that meningeal LVs develop postnatally, appearing first around the foramina in the basal parts of the skull and spinal canal, sprouting along the blood vessels and cranial and spinal nerves to various parts of the meninges surrounding the central nervous system (CNS). VEGF-C, expressed mainly in vascular smooth muscle cells, and VEGFR3 in lymphatic endothelial cells were essential for their development, whereas VEGF-D deletion had no effect. Surprisingly, in adult mice, the LVs showed regression after VEGF-C or VEGFR3 deletion, administration of the tyrosine kinase inhibitor sunitinib, or expression of VEGF-C/D trap, which also compromised the lymphatic drainage function. Conversely, an excess of VEGF-C induced meningeal lymphangiogenesis. The plasticity and regenerative potential of meningeal LVs should allow manipulation of cerebrospinal fluid drainage and neuropathological processes in the CNS.


Assuntos
Vasos Linfáticos/fisiologia , Meninges/fisiologia , Animais , Animais Recém-Nascidos , Transporte Biológico/efeitos dos fármacos , Líquido Cefalorraquidiano/metabolismo , Dependovirus/metabolismo , Deleção de Genes , Humanos , Indóis/farmacologia , Injeções Intraventriculares , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Masculino , Meninges/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Microesferas , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Transdução de Sinais , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiologia , Sunitinibe , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
J Neuroinflammation ; 14(1): 32, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28183352

RESUMO

BACKGROUND: The spatial learning abilities of developing mice benefit from extrinsic cues, such as an enriched environment, with concomitant enhancement in cognitive functions. Interestingly, such enhancements can be further increased through intrinsic Bacillus Calmette-Guérin (BCG) vaccination. RESULTS: Here, we first report that combined neonatal BCG vaccination and exposure to an enriched environment (Enr) induced combined neurobeneficial effects, including hippocampal long-term potentiation, and increased neurogenesis and spatial learning and memory, in mice exposed to the Enr and vaccinated with BCG relative to those in the Enr that did not receive BCG vaccination. Neonatal BCG vaccination markedly induced anti-inflammatory meningeal macrophage polarization both in regular and Enr breeding mice. The meninges are composed of the pia mater, dura mater, and choroid plexus. Alternatively, this anti-inflammatory activity of the meninges occurred simultaneously with increased expression of the neurotrophic factors BDNF/IGF-1 and the M2 microglial phenotype in the hippocampus. Our results reveal a critical role for BCG vaccination in the regulation of neurogenesis and spatial cognition through meningeal macrophage M2 polarization and neurotrophic factor expression; these effects were completely or partially prevented by minocycline or anti-IL-10 antibody treatment, respectively. CONCLUSIONS: Together, we first claim that immunological factor and environmental factor induce a combined effect on neurogenesis and cognition via a common pathway-meningeal macrophage M2 polarization. We also present a novel functional association between peripheral T lymphocytes and meningeal macrophages after evoking adaptive immune responses in the periphery whereby T lymphocytes are recruited to the meninges in response to systemic IFN-γ signaling. This leads to meningeal macrophage M2 polarization, subsequent to microglial M2 activation and neurotrophic factor expression, and eventually promotes a positive behavior.


Assuntos
Vacina BCG/administração & dosagem , Cognição/fisiologia , Meio Ambiente , Macrófagos/metabolismo , Neurogênese/fisiologia , Comportamento Espacial/fisiologia , Animais , Animais Recém-Nascidos , Polaridade Celular/efeitos dos fármacos , Polaridade Celular/fisiologia , Cognição/efeitos dos fármacos , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Meninges/efeitos dos fármacos , Meninges/imunologia , Meninges/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Comportamento Espacial/efeitos dos fármacos
19.
Neuropeptides ; 64: 61-68, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28202186

RESUMO

Primary headaches may be accompanied by increased intracranial blood flow induced by the release of the potent vasodilator calcitonin gene-related peptide (CGRP) from activated meningeal afferents. We aimed to record meningeal and medullary blood flow simultaneously and to localize the sites of CGRP release in rodent preparations in vivo and ex vivo. Blood flow in the exposed rat parietal dura mater and the medulla oblongata was recorded by laser Doppler flowmetry, while the dura was stimulated by topical application of 60mM potassium chloride (KCl). Samples of jugular venous plasma and cerebrospinal fluid (CSF) collected from the cisterna magna were analysed for CGRP concentrations using an enzyme immunoassay. In a hemisected rat skull preparation lined with dura mater the CGRP releasing effect of KCl superfusion was examined. Superfusion of the dura mater with KCl decreased meningeal blood flow unless alpha-adrenoceptors were blocked by phentolamine, whereas the medullary blood flow was increased. The same treatment caused increased CGRP concentrations in jugular plasma and CSF and induced significant CGRP release in the hemisected rat skull preparation. Anaesthesia of the trigeminal ganglion by injection of lidocaine reduced increases in medullary blood flow and CGRP concentration in the CSF upon meningeal KCl application. CGRP release evoked by depolarisation of meningeal afferents is accompanied by increased blood flow in the medulla oblongata but not the dura mater. This discrepancy can be explained by the smooth muscle depolarising effect of KCl and the activation of sympathetic vasoconstrictor mechanisms. The medullary blood flow response is most likely mediated by CGRP released from activated central terminals of trigeminal afferents. Increased blood supply of the medulla oblongata and CGRP release into the CSF may also occur in headaches accompanying vigorous activation of meningeal afferents.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/líquido cefalorraquidiano , Circulação Cerebrovascular/efeitos dos fármacos , Dura-Máter/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Animais , Cefaleia/fisiopatologia , Masculino , Meninges/efeitos dos fármacos , Ratos Wistar , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/efeitos dos fármacos , Gânglio Trigeminal/efeitos dos fármacos
20.
Dev Biol ; 420(1): 148-165, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27671872

RESUMO

Growth and maturation of the cerebrovasculature is a vital event in neocortical development however mechanisms that control cerebrovascular development remain poorly understood. Mutations in or deletions that include the FOXC1 gene are associated with congenital cerebrovascular anomalies and increased stroke risk in patients. Foxc1 mutant mice display severe cerebrovascular hemorrhage at late gestational ages. While these data demonstrate Foxc1 is required for cerebrovascular development, its broad expression in the brain vasculature combined with Foxc1 mutant's complex developmental defects have made it difficult to pinpoint its function(s). Using global and conditional Foxc1 mutants, we find 1) significant cerebrovascular growth defects precede cerebral hemorrhage and 2) expression of Foxc1 in neural crest-derived meninges and brain pericytes, though not endothelial cells, is required for normal cerebrovascular development. We provide evidence that reduced levels of meninges-derived retinoic acid (RA), caused by defects in meninges formation in Foxc1 mutants, is a major contributing factor to the cerebrovascular growth defects in Foxc1 mutants. We provide data that suggests that meninges-derived RA ensures adequate growth of the neocortical vasculature via regulating expression of WNT pathway proteins and neural progenitor derived-VEGF-A. Our findings offer the first evidence for a role of the meninges in brain vascular development and provide new insight into potential causes of cerebrovascular defects in patients with FOXC1 mutations.


Assuntos
Encéfalo/anormalidades , Fatores de Transcrição Forkhead/genética , Meninges/metabolismo , Mutação/genética , Transdução de Sinais , Tretinoína/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Wnt/metabolismo , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Células Cultivadas , Hemorragia Cerebral/patologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Imuno-Histoquímica , Integrases/metabolismo , Meninges/efeitos dos fármacos , Camundongos , Neocórtex/irrigação sanguínea , Neocórtex/embriologia , Neocórtex/patologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...