Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Transl Res ; 176: 69-80, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27164406

RESUMO

Based on results of hematopoietic stem cell transplantation in animal models of stroke, clinical trials with hematopoietic stem cells administered intra-arterially or intravenously have been initiated in patients. Although intra-arterial injection is expected to deliver transplanted cells more directly to the ischemic tissue, the optimal route for enhancing clinical outcomes has not been identified in the setting of stroke. In this study, we compared the therapeutic potential of intra-arterial versus intravenous injection of bone marrow derived-mononuclear cells (BM-MNCs) and CD133-positive (CD133(+)) cells in a murine stroke model. We have found that intra-arterial injection of BM-MNCs exaggerates inflammation with accompanying loss of microvascular structures in poststroke brain and no improvement in cortical function. In contrast, intravenous injection of BM-MNCs did not similarly enhance inflammation and improved cortical function. Our results indicate that the optimal route of cell transplantation can vary with different cell populations and highlight possible issues that might arise with intra-arterial cell administration for acute ischemic cerebrovascular disease.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Acidente Vascular Cerebral/terapia , Antígeno AC133/metabolismo , Administração Intravenosa , Animais , Atrofia , Comportamento Animal , Células da Medula Óssea/citologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Contagem de Células , Modelos Animais de Doenças , Humanos , Injeções Intra-Arteriais , Masculino , Camundongos SCID , Microvasos/patologia , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
2.
Stem Cells Dev ; 24(19): 2207-18, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26176265

RESUMO

The goal of this clinical trial was to assess the feasibility and safety of transplanting autologous bone marrow mononuclear cells into patients suffering severe embolic stroke. Major inclusion criteria included patients with cerebral embolism, age 20-75 years, National Institute of Health Stroke Scale (NIHSS) score displaying improvement of ≤ 5 points during the first 7 days after stroke, and NIHSS score of ≥ 10 on day 7 after stroke. Bone marrow aspiration (25 or 50 mL; N = 6 patients in each case) was performed 7-10 days poststroke, and bone marrow mononuclear cells were administrated intravenously. Mean total transplanted cell numbers were 2.5 × 10(8) and 3.4 × 10(8) cells in the lower and higher dose groups, respectively. No apparent adverse effects of administering bone marrow cells were observed. Compared with the lower dose, patients receiving the higher dose of bone marrow cells displayed a trend toward improved neurologic outcomes. Compared with 1 month after treatment, patients receiving cell therapy displayed a trend toward improved cerebral blood flow and metabolic rate of oxygen consumption 6 months after treatment. In comparison with historical controls, patients receiving cell therapy had significantly better neurologic outcomes. Our results indicated that intravenous transplantation of autologous bone marrow mononuclear cells is safe and feasible. Positive results and trends favoring neurologic recovery and improvement in cerebral blood flow and metabolism by cell therapy underscore the relevance of larger scale randomized controlled trials using this approach.


Assuntos
Transplante de Medula Óssea/métodos , Leucócitos Mononucleares/transplante , Acidente Vascular Cerebral/terapia , Administração Intravenosa , Idoso , Antígenos CD34/sangue , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/efeitos adversos , Circulação Cerebrovascular , Citocinas/sangue , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Contagem de Leucócitos , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Oxigênio/sangue , Oxigênio/metabolismo , Recidiva , Transplante Autólogo , Resultado do Tratamento
3.
J Immunol ; 191(1): 369-77, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729438

RESUMO

Promiscuity of pattern recognition receptors, such as receptor for advanced glycation end products (RAGE), allows for a complex regulatory network controlling inflammation. Scavenging of RAGE ligands by soluble RAGE treatment is effective in reducing delayed-type hypersensitivity (DTH), even in RAGE(-/-) mice by 50% (p < 0.001). This has led to the hypothesis that molecules scavenged by soluble RAGE bind to receptors other than RAGE. This study identifies CD166/ALCAM (ALCAM) as a close structural and functional homolog of RAGE, and it shows that binding of S100B to CD166/ALCAM induces dose- and time-dependent expression of members of the NF-κB family in wild type (WT) and RAGE(-/-) mouse endothelial cells. Blocking CD166/ALCAM expression using small interfering RNA completely inhibited S100B-induced NF-κB activation in RAGE(-/-), but not in WT cells. The in vivo significance of these observations was demonstrated by attenuation of DTH in WT and RAGE(-/-) animals pretreated with CD166/ALCAM small interfering RNA by 50% and 40%, respectively (p < 0.001). Experiments in ALCAM(-/-) animals displayed an only slight reduction of 16% in DTH, explained by compensatory reciprocal upregulation of RAGE in animals devoid of CD166/ALCAM, and vice versa. Consistently, ALCAM(-/-) mice, but not WT mice treated with RAGE small interfering RNA show a 35% reduction in DTH, and ALCAM(-/-) RAGE(-/-) double-knockout mice show a 27% reduction in DTH reaction. Thus, S100B is a proinflammatory cytokine bridging RAGE and CD166/ALCAM downstream effector mechanisms, both being compensatory upregulated after genetic deletion of its counterpart.


Assuntos
Molécula de Adesão de Leucócito Ativado/fisiologia , Antígenos CD/fisiologia , Glicoproteínas/fisiologia , Hipersensibilidade Tardia/imunologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Fatores de Crescimento Neural/fisiologia , Peptídeos/fisiologia , Proteínas S100/fisiologia , Antígeno AC133 , Molécula de Adesão de Leucócito Ativado/química , Animais , Antígenos CD/química , Células Cultivadas , Relação Dose-Resposta Imunológica , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/química , Humanos , Hipersensibilidade Tardia/metabolismo , Hipersensibilidade Tardia/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/química , Peptídeos/antagonistas & inibidores , Peptídeos/química , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/biossíntese , Proteínas S100/química , Relação Estrutura-Atividade , Regulação para Cima/imunologia
4.
Neurosci Res ; 76(3): 163-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23603509

RESUMO

To simulate the clinical and pathologic situation in patients with stroke, as well as to evaluate future potential therapeutic approaches, it is essential to have a highly reproducible model that displays long-term survival. Though a range of rodent models has been employed in the literature, there are questions regarding reproducibility, especially in terms of ischemic zone (i.e., degree of ischemia) and long-term survival. We have developed a highly reproducible stroke model that produces a consistent ischemic zone as a result of direct transient occlusion of the middle cerebral artery (MCA) in CB-17 (CB-17/Icr-+/+Jcl) mice. The model employs a thin monofilament to twist the artery resulting in complete interruption of blood flow. Transient ischemia can be induced for up to 240min and the survival rate at 7 days post-ischemia was more than 60%, even in mice subjected to 240min of transient ischemia resulting in hemorrhagic infarction in most animals. Our method can be used to model several pathologic conditions, such as reversible reperfusion injury, delayed neuronal death, necrotic brain injury and hemorrhagic infarction. We believe this preclinical model provides a step forward for testing future therapeutic approaches applicable to patients with ischemic brain injury.


Assuntos
Isquemia Encefálica , Modelos Animais de Doenças , Traumatismo por Reperfusão , Animais , Isquemia Encefálica/mortalidade , Isquemia Encefálica/patologia , Masculino , Camundongos , Traumatismo por Reperfusão/mortalidade , Traumatismo por Reperfusão/patologia , Reprodutibilidade dos Testes
5.
Nat Med ; 18(6): 926-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22581285

RESUMO

This study establishes a mechanism for metabolic hyperalgesia based on the glycolytic metabolite methylglyoxal. We found that concentrations of plasma methylglyoxal above 600 nM discriminate between diabetes-affected individuals with pain and those without pain. Methylglyoxal depolarizes sensory neurons and induces post-translational modifications of the voltage-gated sodium channel Na(v)1.8, which are associated with increased electrical excitability and facilitated firing of nociceptive neurons, whereas it promotes the slow inactivation of Na(v)1.7. In mice, treatment with methylglyoxal reduces nerve conduction velocity, facilitates neurosecretion of calcitonin gene-related peptide, increases cyclooxygenase-2 (COX-2) expression and evokes thermal and mechanical hyperalgesia. This hyperalgesia is reflected by increased blood flow in brain regions that are involved in pain processing. We also found similar changes in streptozotocin-induced and genetic mouse models of diabetes but not in Na(v)1.8 knockout (Scn10(-/-)) mice. Several strategies that include a methylglyoxal scavenger are effective in reducing methylglyoxal- and diabetes-induced hyperalgesia. This previously undescribed concept of metabolically driven hyperalgesia provides a new basis for the design of therapeutic interventions for painful diabetic neuropathy.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Neuropatias Diabéticas/fisiopatologia , Hiperalgesia/etiologia , Nociceptores/efeitos dos fármacos , Aldeído Pirúvico/farmacologia , Canais de Sódio/fisiologia , Animais , Circulação Cerebrovascular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.8 , Condução Nervosa/efeitos dos fármacos , Nociceptores/fisiologia , Estreptozocina , Tetrodotoxina/farmacologia
6.
J Neurosci ; 31(6): 2313-20, 2011 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21307267

RESUMO

Amyloid-ß (Aß) peptide-binding alcohol dehydrogenase (ABAD), an enzyme present in neuronal mitochondria, exacerbates Aß-induced cell stress. The interaction of ABAD with Aß exacerbates Aß-induced mitochondrial and neuronal dysfunction. Here, we show that inhibition of the ABAD-Aß interaction, using a decoy peptide (DP) in vitro and in vivo, protects against aberrant mitochondrial and neuronal function and improves spatial learning/memory. Intraperitoneal administration of ABAD-DP [fused to the transduction of human immunodeficiency virus 1-transactivator (Tat) protein and linked to the mitochondrial targeting sequence (Mito) (TAT-mito-DP) to transgenic APP mice (Tg mAPP)] blocked formation of ABAD-Aß complex in mitochondria, increased oxygen consumption and enzyme activity associated with the mitochondrial respiratory chain, attenuated mitochondrial oxidative stress, and improved spatial memory. Similar protective effects were observed in Tg mAPP mice overexpressing neuronal ABAD decoy peptide (Tg mAPP/mito-ABAD). Notably, inhibition of the ABAD-Aß interaction significantly reduced mitochondrial Aß accumulation. In parallel, the activity of mitochondrial Aß-degrading enzyme PreP (presequence peptidase) was enhanced in Tg mAPP mitochondria expressing the ABAD decoy peptide. These data indicate that segregating ABAD from Aß protects mitochondria/neurons from Aß toxicity; thus, ABAD-Aß interaction is an important mechanism underlying Aß-mediated mitochondrial and neuronal perturbation. Inhibitors of ABAD-Aß interaction may hold promise as targets for the prevention and treatment of Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/ultraestrutura , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/antagonistas & inibidores , 3-Hidroxiacil-CoA Desidrogenases/farmacologia , 3-Hidroxiacil-CoA Desidrogenases/uso terapêutico , Acetilcolinesterase/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Sítios de Ligação/genética , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Imunoprecipitação/métodos , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/genética , Mutação/genética , Espécies Reativas de Oxigênio/metabolismo , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia
7.
J Cereb Blood Flow Metab ; 31(3): 855-67, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20859292

RESUMO

Circulating bone marrow-derived immature cells, including endothelial progenitor cells, have been implicated in homeostasis of the microvasculature. Decreased levels of circulating endothelial progenitor cells, associated with aging and/or cardiovascular risk factors, correlate with poor clinical outcomes in a range of cardiovascular diseases. Herein, we transplanted bone marrow cells from young stroke-prone spontaneously hypertensive rats (SHR-SP) into aged SHR-SP, the latter not exposed to radiation or chemotherapy. Analysis of recipient peripheral blood 28 days after transplantation revealed that 5% of circulating blood cells were of donor origin. Cerebral infarction was induced on day 30 posttransplantation. Animals transplanted with bone marrow from young SHR-SP displayed an increase in density of the microvasculature in the periinfarction zone, reduced ischemic brain damage and improved neurologic function. In vitro analysis revealed enhanced activation of endothelial nitric oxide synthase and reduced activation p38 microtubule-associated protein (MAP) kinase, the latter associated with endothelial apoptosis, in cultures exposed to bone marrow-derived mononuclear cells from young animals versus cells from aged counterparts. Our findings indicate that partial rejuvenation of bone marrow from aged rats with cells from young animals enhances the response to ischemic injury, potentially at the level of endothelial/vascular activation, providing insight into a novel approach ameliorate chronic vascular diseases.


Assuntos
Envelhecimento , Transplante de Medula Óssea , Isquemia Encefálica/patologia , Encéfalo/patologia , Animais , Vasos Sanguíneos/patologia , Transplante de Medula Óssea/métodos , Isquemia Encefálica/complicações , Isquemia Encefálica/fisiopatologia , Córtex Cerebral/patologia , Citocinas/metabolismo , Suscetibilidade a Doenças , Ativação Enzimática , Hibridização in Situ Fluorescente , Mediadores da Inflamação/metabolismo , Injeções , Masculino , Microcirculação , Sistema Nervoso/fisiopatologia , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Endogâmicos SHR/genética , Pele/lesões , Pele/fisiopatologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Taxa de Sobrevida , Cicatrização , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
J Exp Stroke Transl Med ; 3(1): 28-33, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20865060

RESUMO

In order to evaluate novel stroke therapies, it is essential to utilize a highly reproducible model of focal cerebral ischemia. Though a range of rodent stroke models has been employed in the literature, there are persistent issues regarding reproducibility of the ischemic zone, as there is considerable inter-animal and inter-laboratory variation. We have developed a highly reproducible model of stroke that involves direct electrocoagulation of the MCA in SCID (CB-17/lcr-scid/scidJcl) and CB-17 (CB-17/lcr-+/+Jcl) mice. Using a modification of the Tamura method, our results demonstrate reproducible cortical infarction with high survival in the chronic period (up to 180 days) in SCID and CB-17, but not in C57BL/6, mice. We believe that our preclinical model represents a step forward for testing future therapeutic methods potentially applicable to patients with stroke.

9.
J Neurosci Res ; 88(11): 2385-97, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20623538

RESUMO

Acute inflammation in the poststroke period exacerbates neuronal damage and stimulates reparative mechanisms, including neurogenesis. However, only a small fraction of neural stem/progenitor cells survives. In this report, by using a highly reproducible model of cortical infarction in SCID mice, we examined the effects of immunodeficiency on reduction of brain injury, survival of neural stem/progenitor cells, and functional recovery. Subsequently, the contribution of T lymphocytes to neurogenesis was evaluated in mice depleted for each subset of T lymphocyte. SCID mice revealed the reduced apoptosis and enhanced proliferation of neural stem/progenitor cells induced by cerebral cortex after stroke compared with the immunocompetent wild-type mice. Removal of T lymphocytes, especially the CD4(+) T-cell population, enhanced generation of neural stem/progenitor cells, followed by accelerated functional recovery. In contrast, removal of CD25(+) T cells, a cell population including regulatory T lymphocytes, impaired functional recovery through, at least in part, suppression of neurogenesis. Our findings demonstrate a key role of T lymphocytes in regulation of poststroke neurogenesis and indicate a potential novel strategy for cell therapy in repair of the central nervous system.


Assuntos
Apoptose/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Córtex Cerebral/patologia , Terapia de Imunossupressão , Neurogênese/fisiologia , Neurônios/transplante , Células-Tronco/fisiologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/terapia , Animais , Comportamento Animal/fisiologia , Isquemia Encefálica/patologia , Isquemia Encefálica/terapia , Caspase 3/metabolismo , Morte Celular/fisiologia , Infarto Cerebral/patologia , Infarto Cerebral/psicologia , Infarto Cerebral/terapia , Lateralidade Funcional/fisiologia , Imunocompetência , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Subunidade alfa de Receptor de Interleucina-2/genética , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Neuroglia/patologia , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/imunologia
10.
Brain Res ; 1340: 70-80, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20388500

RESUMO

The beneficial effects of angiotensin II type 1 (AT1) receptor blockers (ARB) in cerebrovascular disease have been shown in clinical trials. However, the effects of ARBs vary based on their unique pharmacologic properties. In this study, we focused on telmisartan, a fat-soluble ARB with selective peroxisome proliferator-activated receptor-gamma (PPAR gamma) agonist activity, and investigated its effects on ischemic injury in cerebral vasculature using murine models of both transient and permanent focal ischemia. Analysis by triphenyltetrazolium-staining revealed that pre-treatment of mice with telmisartan reduced stroke volume 72 h after the transient ischemic insult in a dose-dependent manner, though such treatment did not reduce stroke volume due to permanent ischemia. Transient ischemia induced pro-inflammatory adhesion molecules, such as ICAM-1 and P-selectin in the ischemic region, and treatment with telmisartan diminished the expression of these adhesion molecules with diminished infiltration of inflammatory cells. The beneficial effect of telmisartan was attenuated, in part, by administration of a PPAR gamma antagonist. Treatment with valsartan (an ARB without PPAR gamma agonist activity) also decreased ischemic injury after transient ischemia, though to a lesser extent than telmisartan. Our findings indicate that telmisartan has a beneficial effect in a murine model of ischemia/reperfusion injury through blockade of AT1 receptors, and, in addition, due to a positive effect via its specific anti-inflammatory PPAR gamma agonist activity.


Assuntos
Benzimidazóis/farmacologia , Benzoatos/farmacologia , Isquemia Encefálica/tratamento farmacológico , Infarto Cerebral/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Benzimidazóis/uso terapêutico , Benzoatos/uso terapêutico , Isquemia Encefálica/fisiopatologia , Infarto Cerebral/fisiopatologia , Modelos Animais de Doenças , Gliose/tratamento farmacológico , Gliose/fisiopatologia , Gliose/prevenção & controle , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos SCID , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Receptor Tipo 1 de Angiotensina , Traumatismo por Reperfusão/fisiopatologia , Telmisartan
11.
Eur J Neurosci ; 31(1): 90-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20104652

RESUMO

Increasing evidence points to accelerated neurogenesis after stroke, and support of such endogenous neurogenesis has been shown to improve stroke outcome in experimental animal models. The present study analyses post-stroke cerebral cortex after cardiogenic embolism in autoptic human brain. Induction of nestin- and musashi-1-positive cells, potential neural stem/progenitor cells, was observed at the site of ischemic lesions from day 1 after stroke. These two cell populations were present at distinct locations and displayed different temporal profiles of marker expression. However, no surviving differentiated mature neural cells were observed by 90 days after stroke in the previously ischemic region. Consistent with recent reports of neurogenesis in the cerebral cortex after induction of stroke in rodent models, the present current data indicate the presence of a regional regenerative response in human cerebral cortex. Furthermore, observations underline the potential importance of supporting survival and differentiation of endogenous neural stem/progenitor cells in post-stroke human brain.


Assuntos
Células-Tronco Adultas/fisiologia , Isquemia Encefálica/fisiopatologia , Córtex Cerebral/lesões , Córtex Cerebral/fisiopatologia , Neurônios/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Isquemia Encefálica/patologia , Córtex Cerebral/patologia , Feminino , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Embolia Intracraniana/patologia , Embolia Intracraniana/fisiopatologia , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Nestina , Proteínas de Ligação a RNA/metabolismo , Acidente Vascular Cerebral/patologia , Fatores de Tempo
12.
FASEB J ; 24(4): 1043-55, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19906677

RESUMO

Microglia are critical for amyloid-beta peptide (Abeta)-mediated neuronal perturbation relevant to Alzheimer's disease (AD) pathogenesis. We demonstrate that overexpression of receptor for advanced glycation end products (RAGE) in imbroglio exaggerates neuroinflammation, as evidenced by increased proinflammatory mediator production, Abeta accumulation, impaired learning/memory, and neurotoxicity in an Abeta-rich environment. Transgenic (Tg) mice expressing human mutant APP (mAPP) in neurons and RAGE in microglia displayed enhanced IL-1beta and TNF-alpha production, increased infiltration of microglia and astrocytes, accumulation of Abeta, reduced acetylcholine esterase (AChE) activity, and accelerated deterioration of spatial learning/memory. Notably, introduction of a signal transduction-defective mutant RAGE (DN-RAGE) to microglia attenuates deterioration induced by Abeta. These findings indicate that RAGE signaling in microglia contributes to the pathogenesis of an inflammatory response that ultimately impairs neuronal function and directly affects amyloid accumulation. We conclude that blockade of microglial RAGE may have a beneficial effect on Abeta-mediated neuronal perturbation relevant to AD pathogenesis.-Fang, F., Lue, L.-F., Yan, S., Xu, H., Luddy, J. S., Chen, D., Walker, D. G., Stern, D. M., Yan, S., Schmidt, A. M., Chen, J. X., Yan, S. S. RAGE-dependent signaling in microglia contributes to neuroinflammation, Abeta accumulation, and impaired learning/memory in a mouse model of Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Memória , Microglia/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Modelos Animais de Doenças , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Aprendizagem , Camundongos , Camundongos Transgênicos , Microglia/patologia , Proteínas Quinases Ativadas por Mitógeno , Mutação , Neurônios/metabolismo , Neurônios/patologia , Receptor para Produtos Finais de Glicação Avançada/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
13.
Mol Cell Biochem ; 333(1-2): 9-26, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19585224

RESUMO

Vascular endothelium is vulnerable to the attack of glucose-derived oxoaldehydes (glyoxal and methylglyoxal) during diabetes, through the formation of advanced glycation end products (AGEs). Although aminoguanidine (AG) has been shown to protect against the AGE-induced adverse effects, its protection against the glyoxal-induced alterations in vascular endothelial cells (ECs) such as cytotoxicity, barrier dysfunction, and inhibition of angiogenesis has not been reported and we investigated this in the bovine pulmonary artery ECs (BPAECs). The results showed that glyoxal (1-10 mM) significantly induced cytotoxicity and mitochondrial dysfunction in a dose- and time-dependent (4-12 h) fashion in ECs. Glyoxal was also observed to significantly inhibit EC proliferation. The study also revealed that glyoxal induced EC barrier dysfunction (loss of trans-endothelial electrical resistance), actin cytoskeletal rearrangement, and tight junction alterations in BPAECs. Furthermore, the results revealed that glyoxal significantly inhibited in vitro angiogenesis on the Matrigel. For the first time, this study demonstrated that AG significantly protected against the glyoxal-induced cytotoxicity, barrier dysfunction, cytoskeletal rearrangement, and inhibition of angiogenesis in BPAECs. Therefore, AG appears as a promising protective agent in the treatment of AGE-induced vascular endothelial alterations and dysfunction during diabetes, presumably by blocking the reactivity of the sugar-derived dicarbonyls such as glyoxal and preventing the formation of AGEs.


Assuntos
Citoesqueleto/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Glioxal/farmacologia , Guanidinas/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/patologia , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Células Endoteliais/ultraestrutura , Endotélio Vascular/citologia , Produtos Finais de Glicação Avançada , Guanidinas/uso terapêutico , Óxido Nítrico Sintase/antagonistas & inibidores , Substâncias Protetoras
14.
Proc Natl Acad Sci U S A ; 106(47): 20021-6, 2009 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-19901339

RESUMO

Intracellular amyloid-beta peptide (Abeta) has been implicated in neuronal death associated with Alzheimer's disease. Although Abeta is predominantly secreted into the extracellular space, mechanisms of Abeta transport at the level of the neuronal cell membrane remain to be fully elucidated. We demonstrate that receptor for advanced glycation end products (RAGE) contributes to transport of Abeta from the cell surface to the intracellular space. Mouse cortical neurons exposed to extracellular human Abeta subsequently showed detectable peptide intracellularly in the cytosol and mitochondria by confocal microscope and immunogold electron microscopy. Pretreatment of cultured neurons from wild-type mice with neutralizing antibody to RAGE, and neurons from RAGE knockout mice displayed decreased uptake of Abeta and protection from Abeta-mediated mitochondrial dysfunction. Abeta activated p38 MAPK, but not SAPK/JNK, and then stimulated intracellular uptake of Abeta-RAGE complex. Similar intraneuronal co-localization of Abeta and RAGE was observed in the hippocampus of transgenic mice overexpressing mutant amyloid precursor protein. These findings indicate that RAGE contributes to mechanisms involved in the translocation of Abeta from the extracellular to the intracellular space, thereby enhancing Abeta cytotoxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Peptídeos beta-Amiloides/genética , Animais , Transporte Biológico/fisiologia , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Células Cultivadas , Ativação Enzimática , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Neurônios/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Brain Res ; 1286: 185-91, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19524559

RESUMO

We investigated a non-human primate (NHP) transient global ischemia (TGI) model which was induced by clipping the arteries originating from the aortic arch. Previously we demonstrated that our TGI model in adult Rhesus macaques (Macaca mulatta) results in marked neuronal cell loss in the hippocampal region, specifically the cornu Ammonis (CA1) region. However, we observed varying degrees of hippocampal cell loss among animals. Here, we report for the first time an anomaly of the aortic arch in some Rhesus macaques that appears as a key surgical factor in ensuring the success of the TGI model in this particular NHP. Eleven adult Rhesus macaques underwent the TGI surgery, which involved 10-15-minute clipping of both innominate and subclavian arteries. Animals were allowed to survive between 1 day and 28 days after TGI. Because of our experience and knowledge that Japanese macaques exhibited only innominate and subclavian arteries arising from the aortic arch, macroscopic visualization of these two arteries alone in the Rhesus macaques initially assured us that clipping both arteries was sufficient to produce TGI. During the course of one TGI operation, however, we detected 3 arterial branches arising from the aortic arch, which prompted us to subsequently search for 3 branches in succeeding TGI surgeries. In addition, we performed post-mortem examination of the heart to confirm the number of arterial branches in the aortic arch. Finally, in order to reveal the pathological effect of the aortic arch anomaly, we compared the hippocampal cell loss between animals found to have 3 arterial branches but had all or only two branches clipped during TGI operation. Post-mortem examination revealed that eight NHPs had the typical two arterial aortic branches, but three NHPs displayed an extra arterial aortic branch, indicating that about 30% of Rhesus macaques had 3 arterial branches arising from the aorta. Histological analyses using Nissl staining showed that in NHPs with the aortic arch anomaly clipping only two of three arterial branches led to a partial cell loss and minimal alteration in number of cell layers in the hippocampal region when compared with clipping all three branches, with the hippocampal cell death in the latter resembling the pathological outcome achieved by clipping the two arterial branches in NHPs displaying the typical two-artery aortic arch. The finding that 3 of 11 NHPs exhibited an extra arterial aortic branch recognizes this aortic arch anomaly in Rhesus macaques that warrants a critical surgical maneuver in order to successfully produce consistent TGI-induced hippocampal cell loss.


Assuntos
Aorta Torácica/anormalidades , Modelos Animais de Doenças , Hipocampo/patologia , Ataque Isquêmico Transitório/etiologia , Ataque Isquêmico Transitório/patologia , Macaca mulatta/anormalidades , Animais
16.
Stem Cells ; 27(9): 2185-95, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19557831

RESUMO

Transplantation of neural stem cells (NSCs) has been proposed as a therapy for a range of neurological disorders. To realize the potential of this approach, it is essential to control survival, proliferation, migration, and differentiation of NSCs after transplantation. NSCs are regulated in vivo, at least in part, by their specialized microenvironment or "niche." In the adult central nervous system, neurogenic regions, such as the subventricular and subgranular zones, include NSCs residing in a vascular niche with endothelial cells. Although there is accumulating evidence that endothelial cells promote proliferation of NSCs in vitro, there is no description of their impact on transplanted NSCs. In this study, we grafted cortex-derived stroke-induced neural stem/progenitor cells, obtained from adult mice, onto poststroke cortex in the presence or absence of endothelial cells, and compared survival, proliferation, and neuronal differentiation of the neural precursors in vivo. Cotransplantation of endothelial cells and neural stem/progenitor cells increased survival and proliferation of ischemia-induced neural stem/progenitor cells and also accelerated neuronal differentiation compared with transplantation of neural precursors alone. These data indicate that reconstitution of elements in the vascular niche enhances transplantation of adult neural progenitor cells.


Assuntos
Infarto Cerebral , Células Endoteliais/citologia , Células Endoteliais/transplante , Neurônios/citologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/fisiologia , Células Cultivadas , Imuno-Histoquímica , Masculino , Camundongos
17.
Eur J Neurosci ; 29(9): 1842-52, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19473237

RESUMO

The CNS has the potential to marshal strong reparative mechanisms, including activation of endogenous neurogenesis, after a brain injury such as stroke. However, the response of neural stem/progenitor cells to stroke is poorly understood. Recently, neural stem/progenitor cells have been identified in the cerebral cortex, as well as previously recognized regions such as the subventricular or subgranular zones of the hippocampus, suggesting that a contribution of cortex-derived neural stem/progenitor cells may repair ischemic lesions of the cerebral cortex. In the present study, using a highly reproducible murine model of cortical infarction, we have found nestin-positive cells in the post-stroke cerebral cortex, but not in the non-ischemic cortex. Cells obtained from the ischemic core of the post-stroke cerebral cortex formed neurosphere-like cell clusters expressing nestin; such cells had the capacity for self-renewal and differentiated into electrophysiologically functional neurons, astrocytes and myelin-producing oligodendrocytes. Nestin-positive cells from the stroke-affected cortex migrated into the peri-infarct area and differentiated into glial cells in vivo. Although we could not detect differentiation of nestin-positive cells into neurons in vivo, our current observations indicate that endogenous neural stem/progenitors with the potential to become neurons can develop within post-stroke cerebral cortex.


Assuntos
Córtex Cerebral/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Astrócitos/fisiologia , Diferenciação Celular , Movimento Celular , Células Cultivadas , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurogênese , Neuroglia/fisiologia , Oligodendroglia/fisiologia , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acidente Vascular Cerebral/fisiopatologia
18.
J Alzheimers Dis ; 16(4): 833-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19387116

RESUMO

Receptor for Advanced Glycation Endproducts (RAGE) is a multiligand member of the immunoglobulin superfamily of cell surface molecules which serves as a receptor for amyloid-beta peptide (Abeta) on neurons, microglia, astrocytes, and cells of vessel wall. Increased expression of RAGE is observed in regions of the brain affected by Alzheimer's disease (AD), and Abeta-RAGE interaction in vitro leads to cell stress with the generation of reactive oxygen species and activation of downstream signaling mechanisms including the MAP kinase pathway. RAGE-mediated activation of p38 MAP kinase in neurons causes Abeta-induced inhibition of long-term potentiation in slices of entorhinal cortex. Increased expression of RAGE in an Abeta-rich environment, using transgenic mouse models, accelerates and accentuates pathologic, biochemical, and behavioral abnormalities compared with mice overexpressing only mutant amyloid-beta protein precursor. Interception of Abeta interaction with RAGE, by infusion of soluble RAGE, decreases Abeta content and amyloid load, as well as improving learning/memory and synaptic function, in a murine transgenic model of Abeta accumulation. These data suggest that RAGE may be a therapeutic target for AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Receptores Imunológicos/metabolismo , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Receptor para Produtos Finais de Glicação Avançada
20.
J Cereb Blood Flow Metab ; 29(1): 34-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18698330

RESUMO

Increasing evidence points to a role for circulating endothelial progenitors, including populations of CD34-positive (CD34(+)) cells present in peripheral blood, in vascular homeostasis and neovascularization. In this report, circulating CD34(+) cells in individuals with a history of cerebral infarction were correlated with changes in neurologic function over a period of 1 year. Patients with decreased levels of CD34(+) cells displayed significant worsening in neurologic function, evaluated by the Barthel Index and Clinical Dementia Rating. These results support the hypothesis that levels of circulating CD34(+) cells have prognostic value for neural function, consistent with their potential role in maintaining cerebral circulation.


Assuntos
Antígenos CD34/metabolismo , Movimento Celular , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Idoso , Feminino , Seguimentos , Humanos , Masculino , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...