Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Neuropathology ; 42(2): 134-140, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35037303

RESUMO

A 79-year-old man presented with subacute onset of dementia. Brain magnetic resonance imaging revealed leukoencephalopathy in the posterior lobes with presence of microbleeds. Although clinical manifestation suggested a diagnosis of leukoencephalopathy associated with cerebral amyloid angiopathy (CAA), the patient died of sudden rupture of an aneurysm of the thoracic aorta two months after the onset of dementia. Autopsy revealed pathological features of advanced-stage Alzheimer's disease. Immunohistochemistry for amyloid-ß revealed CAA mainly affecting arteries but not capillaries. Klüver-Barrera staining revealed white matter edema predominantly in the occipital lobes without ischemic changes. Perivascular cuffing was found to be sparse, but there was no evidence of angiitis. Pathological findings suggest that leukoencephalopathy was caused by the disruption of the blood-brain barrier rather than ischemia. Because the present patient died before immunotherapy, his neuropathological findings could reflect the pathomechanism of the acute stage of leukoencephalopathy with CAA.


Assuntos
Doença de Alzheimer , Angiopatia Amiloide Cerebral , Leucoencefalopatias , Substância Branca , Idoso , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Angiopatia Amiloide Cerebral/complicações , Angiopatia Amiloide Cerebral/diagnóstico por imagem , Edema/complicações , Edema/patologia , Humanos , Leucoencefalopatias/patologia , Imageamento por Ressonância Magnética , Masculino , Substância Branca/patologia
2.
Radiol Med ; 127(9): 1032-1045, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35907157

RESUMO

Vessel wall MR imaging (VW-MRI) has been introduced into clinical practice and applied to a variety of diseases, and its usefulness has been reported. High-resolution VW-MRI is essential in the diagnostic workup and provides more information than other routine MR imaging protocols. VW-MRI is useful in assessing lesion location, morphology, and severity. Additional information, such as vessel wall enhancement, which is useful in the differential diagnosis of atherosclerotic disease and vasculitis could be assessed by this special imaging technique. This review describes the VW-MRI technique and its clinical applications in arterial disease, venous disease, vasculitis, and leptomeningeal disease.


Assuntos
Imageamento por Ressonância Magnética , Vasculite , Humanos , Angiografia por Ressonância Magnética/métodos , Imageamento por Ressonância Magnética/métodos
3.
Int J Mol Sci ; 23(4)2022 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-35216347

RESUMO

Oligodendrocyte precursor cells (OPCs) serve as progenitor cells of terminally differentiated oligodendrocytes. Past studies have confirmed the importance of epigenetic system in OPC differentiation to oligodendrocytes. High mobility group A1 (HMGA1) is a small non-histone nuclear protein that binds DNA and modifies the chromatin conformational state. However, it is still completely unknown about the roles of HMGA1 in the process of OPC differentiation. In this study, we prepared primary OPC cultures from the neonatal rat cortex and examined whether the loss- and gain-of-function of HMGA1 would change the mRNA levels of oligodendrocyte markers, such as Cnp, Mbp, Myrf and Plp during the process of OPC differentiation. In our system, the mRNA levels of Cnp, Mbp, Myrf and Plp increased depending on the oligodendrocyte maturation step, but the level of Hmga1 mRNA decreased. When HMGA1 was knocked down by a siRNA approach, the mRNA levels of Cnp, Mbp, Myrf and Plp were smaller in OPCs with Hmga1 siRNA compared to the ones in the control OPCs. On the contrary, when HMGA1 expression was increased by transfection of the Hmga1 plasmid, the mRNA levels of Cnp, Mbp, Myrf and Plp were slightly larger compared to the ones in the control OPCs. These data may suggest that HMGA1 participates in the process of OPC differentiation by regulating the mRNA expression level of myelin-related genes.


Assuntos
Marcadores Genéticos/genética , Proteína HMGA1a/genética , Células Precursoras de Oligodendrócitos/metabolismo , Transcrição Gênica/genética , Animais , Diferenciação Celular/genética , Bainha de Mielina/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Células-Tronco/metabolismo
4.
J Stroke Cerebrovasc Dis ; 31(6): 106442, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35305535

RESUMO

OBJECTIVES: To evaluate longitudinally the muscle properties of acute stroke patients and examine the association between physical activity and nutritional intake. MATERIALS AND METHODS: This study enrolled 21 stroke patients (72.7±10.4 years). Muscle quantity (fat-free mass, appendicular skeletal muscle mass) and quality (extracellular water/intracellular water ratio, phase angle) were assessed using a bioelectrical impedance device at baseline (within three days) and two weeks after stroke onset. Physical activity and sedentary were calculated from the accelerometer data. Total energy and protein intake were calculated from the dietary surveys as nutritional intake. The association of physical activity, sedentary, and nutritional intake with the rate of changes in muscle properties was examined. RESULTS: The fat-free mass significantly decreased (from 43.4±8.0 to 42.2±7.6 kg), and the skeletal muscle was unchanged (from 17.8±4.2 to 17.7±4.0 kg) after two weeks. The extracellular water/intracellular water ratio significantly increased (from 0.63±0.02 to 0.65±0.03) and the phase angle significantly decreased (from 5.1±0.6 to 4.9±0.8°), suggesting that the muscle quality have declined. Correlation analysis showed that the extracellular water/intracellular water ratio was significantly associated with physical activity [metabolic equivalents (ρ=-0.61)] and sedentary (ρ=0.67) and that the phase angle was significantly associated with physical activity [metabolic equivalents (ρ=0.69)], sedentary (ρ=-0.68), and nutritional intake [total energy (r=0.45), protein (r=0.45)]. CONCLUSIONS: The fat-free mass and muscle quality (extracellular water/intracellular water ratio and phase angle) declined two weeks after stroke. Physical activity and nutritional intake were lower in patients with decreased muscle quality, suggesting the importance of exercise and nutrition in the acute phase.


Assuntos
Composição Corporal , Acidente Vascular Cerebral , Composição Corporal/fisiologia , Ingestão de Alimentos , Exercício Físico , Humanos , Músculo Esquelético , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/terapia , Água
5.
Angiogenesis ; 24(1): 35-46, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32918673

RESUMO

Blood-brain barrier (BBB) dysfunction underlies the pathogenesis of many neurological diseases. Platelet-derived growth factor receptor-alpha (PDGFRα) induces hemorrhagic transformation (HT) downstream of tissue plasminogen activator in thrombolytic therapy of acute stroke. Thus, PDGFs are attractive therapeutic targets for BBB dysfunction. In the present study, we examined the role of PDGF signaling in the process of tissue remodeling after middle cerebral arterial occlusion (MCAO) in mice. Firstly, we found that imatinib increased lesion size after permanent MCAO in wild-type mice. Moreover, imatinib-induced HT only when administrated in the subacute phase of MCAO, but not in the acute phase. Secondly, we generated genetically mutated mice (C-KO mice) that showed decreased expression of perivascular PDGFRα. Additionally, transient MCAO experiments were performed in these mice. We found that the ischemic lesion size was not affected; however, the recruitment of PDGFRα/type I collagen-expressing perivascular cells was significantly downregulated, and HT and IgG leakage was augmented only in the subacute phase of stroke in C-KO mice. In both experiments, we found that the expression of tight junction proteins and PDGFRß-expressing pericyte coverage was not significantly affected in imatinib-treated mice and in C-KO mice. The specific implication of PDGFRα signaling was suggestive of protective effects against BBB dysfunction during the subacute phase of stroke. Vascular TGF-ß1 expression was downregulated in both imatinib-treated and C-KO mice, along with sustained levels of MMP9. Therefore, PDGFRα effects may be mediated by TGF-ß1 which exerts potent protective effects in the BBB.


Assuntos
Vasos Sanguíneos/metabolismo , Barreira Hematoencefálica/fisiopatologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Acidente Vascular Cerebral/complicações , Animais , Colágeno Tipo I/metabolismo , Hemorragia/patologia , Mesilato de Imatinib , Imunoglobulina G/metabolismo , Infarto da Artéria Cerebral Média/complicações , AVC Isquêmico/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Knockout , Fator de Crescimento Transformador beta1/metabolismo
6.
Int J Mol Sci ; 21(23)2020 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-33260683

RESUMO

A-kinase anchor protein 12 (AKAP12) is a scaffolding protein that associates with intracellular molecules to regulate multiple signal transductions. Although the roles of AKAP12 in the central nervous system are still relatively understudied, it was previously shown that AKAP12 regulates blood-retinal barrier formation. In this study, we asked whether AKAP12 also supports the function and integrity of the blood-brain barrier (BBB). In a mouse model of focal ischemia, the expression level of AKAP12 in cerebral endothelial cells was upregulated during the acute phase of stroke. Also, in cultured cerebral endothelial cells, oxygen-glucose deprivation induced the upregulation of AKAP12. When AKAP12 expression was suppressed by an siRNA approach in cultured endothelial cells, endothelial permeability was increased along with the dysregulation of ZO-1/Claudin 5 expression. In addition, the loss of AKAP12 expression caused an upregulation/activation of the Rho kinase pathway, and treatment of Rho kinase inhibitor Y-27632 mitigated the increase of endothelial permeability in AKAP12-deficient endothelial cell cultures. These in vitro findings were confirmed by our in vivo experiments using Akap12 knockout mice. Compared to wild-type mice, Akap12 knockout mice showed a larger extent of BBB damage after stroke. However, the inhibition of rho kinase by Y-27632 tightened the BBB in Akap12 knockout mice. These data may suggest that endogenous AKAP12 works to alleviate the damage and dysfunction of the BBB caused by ischemic stress. Therefore, the AKAP12-rho-kinase signaling pathway represents a novel therapeutic target for stroke.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Proteínas de Ciclo Celular/metabolismo , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Animais , Permeabilidade da Membrana Celular , Endotélio Vascular/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quinases Associadas a rho/metabolismo
7.
Glia ; 67(4): 718-728, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30793389

RESUMO

During development or after brain injury, oligodendrocyte precursor cells (OPCs) differentiate into oligodendrocytes to supplement the number of oligodendrocytes. Although mechanisms of OPC differentiation have been extensively examined, the role of epigenetic regulators, such as histone deacetylases (HDACs) and DNA methyltransferase enzymes (DNMTs), in this process is still mostly unknown. Here, we report the differential roles of epigenetic regulators in OPC differentiation. We prepared primary OPC cultures from neonatal rat cortex. Our cultured OPCs expressed substantial amounts of mRNA for HDAC1, HDAC2, DNMT1, and DNMT3a. mRNA levels of HDAC1 and HDAC2 were both decreased by the time OPCs differentiated into myelin-basic-protein expressing oligodendrocytes. However, DNMT1 or DNMT3a mRNA level gradually decreased or increased during the differentiation step, respectively. We then knocked down those regulators in cultured OPCs with siRNA technique before starting OPC differentiation. While HDAC1 knockdown suppressed OPC differentiation, HDAC2 knockdown promoted OPC differentiation. DNMT1 knockdown also suppressed OPC differentiation, but unlike HDAC1/2, DNMT1-deficient cells showed cell damage during the later phase of OPC differentiation. On the other hand, when OPCs were transfected with siRNA for DNMT3a, the number of OPCs was decreased, indicating that DNMT3a may participate in OPC survival/proliferation. Taken together, these data demonstrate that each epigenetic regulator has different phase-specific roles in OPC survival and differentiation.


Assuntos
Epigênese Genética/fisiologia , Células Precursoras de Oligodendrócitos/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Córtex Cerebral/citologia , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , DNA Metiltransferase 3A , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transfecção
8.
J Neurosci Res ; 97(7): 828-845, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30891830

RESUMO

Oligodendrocytes (OLs) show heterogeneous properties that depend on their location in the central nervous system (CNS). In this regard, the investigation of oligodendrocyte precursor cells (OPCs) derived from human pluripotent stem cells (hPSCs) should be reconsidered, particularly in cases of brain-predominant disorders for which brain-derived OPCs are more appropriate than spinal cord-derived OPCs. Furthermore, animal-derived components are responsible for culture variability in the derivation and complicate clinical translation. In the present study, we established a xeno-free system to induce forebrain OPCs from hPSCs. We induced human forebrain neural stem cells (NSCs) on Laminin 511-E8 and directed the differentiation to the developmental pathway for forebrain OLs with SHH and FGF signaling. OPCs were characterized by the expression of OLIG2, NKX2.2, SOX10, and PDGFRA, and subsequent maturation into O4+ cells. In vitro characterization showed that >85% of the forebrain OPCs (O4+ ) underwent maturation into OLs (MBP+ ) 3 weeks after mitogen removal. Upon intracranial transplantation, the OPCs survived, dispersed in the corpus callosum, and matured into (GSTπ+ ) OLs in the host brains 3 months after transplantation. These findings suggest our xeno-free induction of forebrain OPCs from hPSCs could accelerate clinical translation for brain-specific disorders.


Assuntos
Células Precursoras de Oligodendrócitos/transplante , Células-Tronco Pluripotentes/metabolismo , Prosencéfalo/transplante , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Linhagem Celular , Expressão Gênica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Células-Tronco Neurais/metabolismo , Proteínas Nucleares , Células Precursoras de Oligodendrócitos/metabolismo , Prosencéfalo/metabolismo , Ratos , Fatores de Transcrição
9.
Stem Cells ; 36(5): 751-760, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29314444

RESUMO

Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes in cerebral white matter. However, the underlying mechanisms that regulate this process remain to be fully defined, especially in adult brains. Recently, it has been suggested that signaling via A-kinase anchor protein 12 (AKAP12), a scaffolding protein that associates with intracellular molecules such as protein kinase A, may be involved in Schwann cell homeostasis and peripheral myelination. Here, we asked whether AKAP12 also regulates the mechanisms of myelination in the CNS. AKAP12 knockout mice were compared against wild-type (WT) mice in a series of neurochemical and behavioral assays. Compared with WTs, 2-months old AKAP12 knockout mice exhibited loss of myelin in white matter of the corpus callosum, along with perturbations in working memory as measured by a standard Y-maze test. Unexpectedly, very few OPCs expressed AKAP12 in the corpus callosum region. Instead, pericytes appeared to be one of the major AKAP12-expressing cells. In a cell culture model system, conditioned culture media from normal pericytes promoted in-vitro OPC maturation. However, conditioned media from AKAP12-deficient pericytes did not support the OPC function. These findings suggest that AKAP12 signaling in pericytes may be required for OPC-to-oligodendrocyte renewal to maintain the white matter homeostasis in adult brain. Stem Cells 2018;36:751-760.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Neurais/citologia , Oligodendroglia/metabolismo , Substância Branca/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Envelhecimento , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células/fisiologia , Células Cultivadas , Meios de Cultivo Condicionados , Camundongos Knockout , Bainha de Mielina/metabolismo , Neurogênese/fisiologia , Oligodendroglia/citologia , Substância Branca/citologia
10.
Int J Mol Sci ; 20(10)2019 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-31126115

RESUMO

Vascular risk factors, such as type 2 diabetes mellitus (T2DM), are associated with the increased risk of Alzheimer's disease. One of the common T2DM medications, dipeptidyl peptidase (DPP)-4 inhibitors, have a minimum risk for hypoglycemia and have recently been suggested to ameliorate ß-amyloid pathology. However, conflicting results have been reported regarding the effects of DPP-4 inhibition on cognitive function and tau pathology. Thus, we investigated whether inhibiting DPP-4 affects tau pathology and cognition in a mouse model of tauopathy with hyperglycemia. Male mice overexpressing the P301S mutant human microtubule-associated protein tau gene (PS19) were fed either a low or high-fat diet. PS19 mice were then administered either linagliptin, a DPP-4 inhibitor, or vehicle, from 6 weeks to 8 months of age. Linagliptin-treated mice exhibited higher levels of glucagon-like peptide-1 and decreased fasting blood glucose, compared with the vehicle-treated mice at 8 months. Linagliptin treatment significantly restored spatial reference memory and increased cerebral blood flow without affecting phosphorylation levels of tau or endothelial nitric oxide synthase (eNOS) in the brain. Linagliptin may ameliorate HFD-induced cognitive worsening in tauopathy, at least partially, by increasing cerebral perfusion via the eNOS-independent pathway.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Hiperglicemia/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Linagliptina/uso terapêutico , Tauopatias/tratamento farmacológico , Animais , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Humanos , Hiperglicemia/complicações , Hiperglicemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tauopatias/complicações , Tauopatias/patologia
11.
Stroke ; 49(4): 1003-1010, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29511131

RESUMO

BACKGROUND AND PURPOSE: Endothelial progenitor cells (EPCs) have been extensively investigated as a therapeutic approach for repairing the vascular system in cerebrovascular diseases. Beyond vascular regeneration per se, EPCs may also release factors that affect the entire neurovascular unit. Here, we aim to study the effects of the EPC secretome on oligovascular remodeling in a mouse model of white matter injury after prolonged cerebral hypoperfusion. METHODS: The secretome of mouse EPCs was analyzed with a proteome array. In vitro, the effects of the EPC secretome and its factor angiogenin were assessed on primary oligodendrocyte precursor cells and mature human cerebral microvascular endothelial cells (hCMED/D3). In vivo, mice were subjected to permanent bilateral common carotid artery stenosis, then treated with EPC secretome at 24 hours and at 1 week, and cognitive outcome was evaluated with the Y maze test together with oligodendrocyte precursor cell proliferation/differentiation and vascular density in white matter at 4 weeks. RESULTS: Multiple growth factors, cytokines, and proteases were identified in the EPC secretome, including angiogenin. In vitro, the EPC secretome significantly enhanced endothelial and oligodendrocyte precursor cell proliferation and potentiated oligodendrocyte precursor cell maturation. Angiogenin was proved to be a key factor since pharmacological blockade of angiogenin signaling negated the positive effects of the EPC secretome. In vivo, treatment with the EPC secretome increased vascular density, myelin, and mature oligodendrocytes in white matter and rescued cognitive function in the mouse hypoperfusion model. CONCLUSIONS: Factors secreted by EPCs may ameliorate white matter damage in the brain by boosting oligovascular remodeling.


Assuntos
Indutores da Angiogênese/farmacologia , Estenose das Carótidas/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Ribonuclease Pancreático/farmacologia , Remodelação Vascular/efeitos dos fármacos , Substância Branca/efeitos dos fármacos , Animais , Isquemia Encefálica/metabolismo , Meios de Cultivo Condicionados , Citocinas/metabolismo , Modelos Animais de Doenças , Glutationa S-Transferase pi/metabolismo , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Proteína Básica da Mielina/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Peptídeo Hidrolases/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Ribonuclease Pancreático/metabolismo , Substância Branca/irrigação sanguínea
12.
J Neurosci Res ; 96(5): 863-874, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29110329

RESUMO

The use of human induced pluripotent stem cells (hiPSCs) eliminates the ethical issues associated with fetal or embryonic materials, thus allowing progress in cell therapy research for ischemic stroke. Strict regulation of cell therapy development requires the xeno-free condition to eliminate clinical complications. Maintenance of hiPSCs with feeder-free condition presents a higher degree of spontaneous differentiation in comparison with conventional cultures. Therefore, feeder-free derivation might be not ideal for developing transplantable hiPSC derivatives. We developed the feeder-free condition for differentiation of cortical neurons from hiPSCs. Then, we evaluated the cells' characteristics upon transplantation into the sham and focal brain ischemia on adult male Wistar rats. Grafts in lesioned brains demonstrated polarized reactivity toward the ischemic border, indicated by directional preferences in axonal outgrowth and cellular migration, with no influence on graft survival. Following the transplantation, forelimb asymmetry was better restored compared with controls. Herein, we provide evidence to support the use of the xeno-free condition for the development of cell therapy for ischemic stroke.


Assuntos
Isquemia Encefálica/terapia , Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Animais , Isquemia Encefálica/patologia , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Células-Tronco Neurais/citologia , Neurônios/citologia , Distribuição Aleatória , Ratos , Ratos Wistar
13.
Microbiol Immunol ; 2018 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-29771464

RESUMO

Accumulation of amyloid ß (Aß40 and Aß42) in the brain is a characteristic of Alzheimer's disease (AD). Because neprilysin (NEP) is a major Aß-degrading enzyme, NEP delivery in the brain is a promising gene therapy for AD. Borna disease virus (BoDV) vector enables long-term transduction of foreign genes in the central nerve system. Here, we evaluated the proteolytic ability of NEP transduced by the BoDV vector and found that the amounts of Aß40 and Aß42 significantly decreased, which suggests that NEP expressed from the BoDV vector is functional to degrade Aß.

14.
Dement Geriatr Cogn Disord ; 44(5-6): 343-353, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29421784

RESUMO

BACKGROUND: Cerebral amyloid angiopathy (CAA) is classified as type 1 with capillary amyloid ß (Aß) or type 2 without capillary Aß. While it is known that CAA activates complement, an inflammatory mediator, there is no information on the relationship between capillary Aß and complement activation. METHODS: We evaluated 34 autopsy brains, including 22 with CAA and 12 with other neurodegenerative diseases. We assessed the vascular density of CAA by analyzing the expression of complement (C1q, C3d, C6, C5b-9), macrophage scavenger receptor (MSR), and apolipoprotein E (ApoE). RESULTS: Capillary immunostaining for C1q, C3d, MSR, and ApoE was identified almost exclusively in CAA-type1 brains. There was intense expression of C1q, C3d, MSR, and ApoE, as well as weaker expression of C5b-9 and C6 in the arteries/ arterioles of both CAA subtypes, but not in control brains. C5b-9 and C6 were preferentially expressed in arteries/arterioles with subcortical hemorrhage or cortical superficial siderosis. Triple immunofluorescence revealed that C1q, C3d, and ApoE were colocalized with Aß in CAA brain capillaries. CONCLUSION: Complement, MSR, and ApoE were only coexpressed in the presence of Aß accumulation in capillaries, suggesting a role for complement activation in the propagation of Aß. Additionally, C5b-9 expression may be associated with hemorrhagic brain injury in CAA.


Assuntos
Capilares/patologia , Angiopatia Amiloide Cerebral/patologia , Ativação do Complemento , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Arteríolas/metabolismo , Arteríolas/patologia , Autopsia , Encéfalo/patologia , Capilares/metabolismo , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/metabolismo , Proteínas do Sistema Complemento/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Receptores Depuradores/metabolismo
15.
J Neurosci ; 35(41): 14002-8, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26468200

RESUMO

Oligodendrocyte precursor cells (OPCs) in the adult brain contribute to white matter homeostasis. After white matter damage, OPCs compensate for oligodendrocyte loss by differentiating into mature oligodendrocytes. However, the underlying mechanisms remain to be fully defined. Here, we test the hypothesis that, during endogenous recovery from white matter ischemic injury, astrocytes support the maturation of OPCs by secreting brain-derived neurotrophic factor (BDNF). For in vitro experiments, cultured primary OPCs and astrocytes were prepared from postnatal day 2 rat cortex. When OPCs were subjected to chemical hypoxic stress by exposing them to sublethal CoCl2 for 7 d, in vitro OPC differentiation into oligodendrocytes was significantly suppressed. Conditioned medium from astrocytes (astro-medium) restored the process of OPC maturation even under the stressed conditions. When astro-medium was filtered with TrkB-Fc to remove BDNF, the BDNF-deficient astro-medium no longer supported OPC maturation. For in vivo experiments, we analyzed a transgenic mouse line (GFAP(cre)/BDNF(wt/fl)) in which BDNF expression is downregulated specifically in GFAP(+) astrocytes. Both wild-type (GFAP(wt)/BDNF(wt/fl) mice) and transgenic mice were subjected to prolonged cerebral hypoperfusion by bilateral common carotid artery stenosis. As expected, compared with wild-type mice, the transgenic mice exhibited a lower number of newly generated oligodendrocytes and larger white matter damage. Together, these findings demonstrate that, during endogenous recovery from white matter damage, astrocytes may promote oligodendrogenesis by secreting BDNF. SIGNIFICANCE STATEMENT: The repair of white matter after brain injury and neurodegeneration remains a tremendous hurdle for a wide spectrum of CNS disorders. One potentially important opportunity may reside in the response of residual oligodendrocyte precursor cells (OPCs). OPCs may serve as a back-up for generating mature oligodendrocytes in damaged white matter. However, the underlying mechanisms are still mostly unknown. Here, we use a combination of cell biology and an animal model to report a new pathway in which astrocyte-derived BDNF supports oligodendrogenesis and regeneration after white matter damage. These findings provide new mechanistic insight into white matter physiology and pathophysiology, which would be broadly and clinically applicable to CNS disease.


Assuntos
Astrócitos/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diferenciação Celular/fisiologia , Leucoencefalopatias/patologia , Animais , Antimutagênicos/farmacologia , Astrócitos/química , Astrócitos/metabolismo , Isquemia Encefálica/complicações , Fator Neurotrófico Derivado do Encéfalo/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Cromonas/farmacologia , Cobalto/farmacologia , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glutationa S-Transferase pi/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Leucoencefalopatias/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfolinas/farmacologia , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Fosfopiruvato Hidratase/metabolismo , Células-Tronco/fisiologia
16.
Stroke ; 47(4): 1094-100, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26965847

RESUMO

BACKGROUND AND PURPOSE: Pentraxin 3 (PTX3) is released on inflammatory responses in many organs. However, roles of PTX3 in brain are still mostly unknown. Here we asked whether and how PTX3 contributes to blood-brain barrier dysfunction during the acute phase of ischemic stroke. METHODS: In vivo, spontaneously hypertensive rats were subjected to focal cerebral ischemia by transient middle cerebral artery occlusion. At day 3, brains were analyzed to evaluate the cellular origin of PTX3 expression. Correlations with blood-brain barrier breakdown were assessed by IgG staining. In vitro, rat primary astrocytes and rat brain endothelial RBE.4 cells were cultured to study the role of astrocyte-derived PTX3 on vascular endothelial growth factor-mediated endothelial permeability. RESULTS: During the acute phase of stroke, reactive astrocytes in the peri-infarct area expressed PTX3. There was negative correlation between gradients of IgG leakage and PTX3-positive astrocytes. Cell culture experiments showed that astrocyte-conditioned media increased levels of tight junction proteins and reduced endothelial permeability under normal conditions. Removing PTX3 from astrocyte-conditioned media by immunoprecipitation increased endothelial permeability. PTX3 strongly bound vascular endothelial growth factor in vitro and was able to decrease vascular endothelial growth factor-induced endothelial permeability. CONCLUSIONS: Astrocytes in peri-infarct areas upregulate PTX3, which may support blood-brain barrier integrity by regulating vascular endothelial growth factor-related mechanisms. This response in astrocytes may comprise a compensatory mechanism for maintaining blood-brain barrier function after ischemic stroke.


Assuntos
Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Proteína C-Reativa/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Componente Amiloide P Sérico/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular , Meios de Cultivo Condicionados , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Stroke ; 46(9): 2691-4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26243221

RESUMO

BACKGROUND AND PURPOSE: Postconditioning may be a clinically feasible way to protect the brain after a stroke. However, its effects during the recovery phase post stroke remain to be fully elucidated. Here, we examine the hypothesis that ischemic postconditioning amplifies neurogenesis and angiogenesis during stroke recovery. METHODS: Male Sprague-Dawley rats were subjected to 100-minute transient middle cerebral artery occlusion (MCAO) or postconditioning (100-minute middle cerebral artery occlusion plus 10-minute reperfusion plus 10-minute reocclusion). After 2 weeks, infarct volumes, behavioral outcomes, and immunohistochemical markers of neurogenesis and angiogenesis were quantified. RESULTS: Postconditioning significantly reduced infarction and improved neurological outcomes. Concomitantly, brains subjected to postconditioning showed an increase in doublecortin/BrdU and collagen-IV/Ki67-positive cells. CONCLUSIONS: These results suggest that therapeutic effects of postconditioning may involve the promotion of neurogenesis and angiogenic remodeling during the recovery phase after focal cerebral ischemia.


Assuntos
Infarto da Artéria Cerebral Média/terapia , Pós-Condicionamento Isquêmico/métodos , Neovascularização Fisiológica/fisiologia , Neurogênese/fisiologia , Recuperação de Função Fisiológica/fisiologia , Reperfusão/métodos , Animais , Proteína Duplacortina , Masculino , Ratos , Ratos Sprague-Dawley
18.
Hum Mol Genet ; 22(12): 2423-34, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23449626

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by selective dopaminergic cell loss in the substantia nigra, but its pathogenesis remains unclear. The recessively inherited familial PD genes PARK2 and PARK6 have been attributed to mutations in the Parkin and PTEN-induced kinase 1 (PINK1) genes, respectively. Recent reports suggest that PINK1 works upstream of Parkin in the same pathway to regulate mitochondrial dynamics and/or conduct autophagic clearance of damaged mitochondria. This phenomenon is preserved from Drosophila to human cell lines but has not been demonstrated in a vertebrate animal model in vivo. Here, we developed a medaka fish (Oryzias latipes) model that is deficient in Pink1 and Parkin. We found that despite the lack of a conspicuous phenotype in single mutants for Pink1 or Parkin, medaka that are deficient in both genes developed phenotypes similar to that of human PD: late-onset locomotor dysfunction, a decrease in dopamine levels and a selective degeneration of dopaminergic neurons. Further analysis also revealed defects in mitochondrial enzymatic activity as well as cell death. Consistently, PINK1 and Parkin double-deficient MEF showed a further decrease in mitochondrial membrane potential and mitochondrial complex I activity as well as apoptosis compared with single-deficient MEF. Interestingly, these mitochondrial abnormalities in Parkin-deficient MEF were compensated by exogenous PINK1, but not by disease-related mutants. These results suggest that PINK1 and Parkin work in a complementary way to protect dopaminergic neurons by maintaining mitochondrial function in vertebrates.


Assuntos
Dopamina/metabolismo , Proteínas de Peixes/metabolismo , Neurônios/metabolismo , Oryzias/metabolismo , Doença de Parkinson/metabolismo , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Drosophila , Proteínas de Peixes/genética , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neurônios/citologia , Oryzias/genética , Doença de Parkinson/genética , Fenótipo , Proteínas Quinases/genética , Ubiquitina-Proteína Ligases/genética , Vertebrados/genética , Vertebrados/metabolismo
19.
Stroke ; 45(11): 3403-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25213338

RESUMO

BACKGROUND AND PURPOSE: Silent information regulator 2 homolog 1 (SIRT1) is a protein deacetylase that has been reported to suppress neurodegenerative and cardiovascular diseases in model organisms. We hypothesized that neurovascular protection is one of the diverse actions of SIRT1. This study was designed to determine whether SIRT1 protects against the consequences of cerebral hypoperfusion in vivo. METHODS: Sirt1-overexpressing (Sirt1-Tg) mice driven by a prion promoter and their wild-type littermates were subjected to bilateral common carotid artery stenosis using external microcoils. Using Sirt1-Tg mice, we assessed the effect of SIRT1 on cerebral blood flow, cerebral angioarchitecture, histological and ultrastructural changes, and spatial working memory at several time points. We also evaluated the effects of preadministration of SIRT1 inhibitors or endothelial nitric oxide synthase inhibitors on cerebral blood flow after bilateral common carotid artery stenosis in Sirt1-Tg mice. Levels of acetylated and nonacetylated endothelial nitric oxide synthase were measured semiquantitatively with immunoblotting. RESULTS: Cerebral hypoperfusion induced by bilateral common carotid artery stenosis caused memory impairment and histological changes in wild-type littermates. However, these phenotypes were rescued in Sirt1-Tg mice, where cerebral blood flow was maintained even poststenosis. Electron microscopic analyses showed irregularities in the vascular endothelia, such as tight junction openings in wild-type mice, which were absent in Sirt1-Tg littermates. Brain endothelial nitric oxide synthase was acetylated after cerebral hypoperfusion in wild-type littermates but remained unacetylated in Sirt1-Tg mice. Moreover, treatment with SIRT1 inhibitors and endothelial nitric oxide synthase inhibitors abolished the vasculoprotective effects of SIRT1. CONCLUSIONS: Our results indicate that neurovascular endothelial SIRT1 potentiation upregulates the nitric oxide system and counters cerebral hypoperfusion injury. This novel cerebral blood flow-preserving mechanism offers potential molecular targets for future therapeutic intervention.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/prevenção & controle , Circulação Cerebrovascular/fisiologia , Óxido Nítrico Sintase Tipo III/biossíntese , Sirtuína 1/biossíntese , Acetilação , Animais , Lesões Encefálicas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
20.
Neurosci Lett ; 833: 137829, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38788796

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that has been reported to be affected by inflammatory cells, such as microglia and macrophages, through the concept of non-cell autonomous neuronal death. Resident microglia in the human brain and monocyte-derived macrophages (MoDM) infiltrating in tissues are difficult to distinguish. Therefore, the effects of microglia and MoDMs in ALS remain poorly understood. This study aimed to investigate the role of resident microglia and MoDMs in the pathogenesis of ALS using postmortem brain and spinal cord samples. The samples used for immunohistochemical analysis included 11 cases of sporadic ALS and 11 age-matched controls. We stained the cells with TMEM119 to detect resident microglia and CCR2 to detect MoDMs. In ALS cases, TMEM119-immunopositive resident microglia were abundant in the motor cortex and subcortical white matter (SWM) of the motor area, whereas CCR2-immunopositive MoDM was similar to control cases. In addition, the mean density of CD68-immunopositive cells in the SWM significantly correlated with the mean density of pTDP-43-positive GCIs. These results suggest that resident microglial activation plays an important role in the cerebral pathogenesis of ALS and may provide novel therapeutic strategies to target excessive activation of resident microglia in ALS.


Assuntos
Esclerose Lateral Amiotrófica , Encéfalo , Proteínas de Membrana , Microglia , Humanos , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Microglia/metabolismo , Microglia/patologia , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Proteínas de Membrana/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Receptores CCR2/metabolismo , Substância Branca/patologia , Substância Branca/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Idoso de 80 Anos ou mais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA