Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 10(4): 4209-18, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-26967254

RESUMO

The blood-brain barrier (BBB) is partially disrupted in brain tumors. Despite the gaps in the BBB, there is an inadequate amount of pharmacological agents delivered into the brain. Thus, the low delivery efficiency renders many of these agents ineffective in treating brain cancer. In this report, we proposed an "autocatalytic" approach for increasing the transport of nanoparticles into the brain. In this strategy, a small number of nanoparticles enter into the brain via transcytosis or through the BBB gaps. After penetrating the BBB, the nanoparticles release BBB modulators, which enables more nanoparticles to be transported, creating a positive feedback loop for increased delivery. Specifically, we demonstrated that these autocatalytic brain tumor-targeting poly(amine-co-ester) terpolymer nanoparticles (ABTT NPs) can readily cross the BBB and preferentially accumulate in brain tumors at a concentration of 4.3- and 94.0-fold greater than that in the liver and in brain regions without tumors, respectively. We further demonstrated that ABTT NPs were capable of mediating brain cancer gene therapy and chemotherapy. Our results suggest ABTT NPs can prime the brain to increase the systemic delivery of therapeutics for treating brain malignancies.


Assuntos
Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/terapia , Nanopartículas/química , Animais , Antineoplásicos/administração & dosagem , Transporte Biológico , Linhagem Celular Tumoral , Ácidos Decanoicos/química , Sistemas de Liberação de Medicamentos , Etanolaminas/química , Feminino , Terapia Genética , Xenoenxertos , Humanos , Metaloproteinase 2 da Matriz/química , Camundongos , Camundongos Endogâmicos C57BL , Imagem Óptica , Paclitaxel/administração & dosagem , Permeabilidade , Polímeros/química , Purinas/química , Pirazóis/química , Venenos de Escorpião/química , Transcitose , Microambiente Tumoral
2.
Neuron ; 87(1): 95-110, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26119027

RESUMO

The precise regulation of cerebral blood flow is critical for normal brain function, and its disruption underlies many neuropathologies. The extent to which smooth muscle-covered arterioles or pericyte-covered capillaries control vasomotion during neurovascular coupling remains controversial. We found that capillary pericytes in mice and humans do not express smooth muscle actin and are morphologically and functionally distinct from adjacent precapillary smooth muscle cells (SMCs). Using optical imaging we investigated blood flow regulation at various sites on the vascular tree in living mice. Optogenetic, whisker stimulation, or cortical spreading depolarization caused microvascular diameter or flow changes in SMC but not pericyte-covered microvessels. During early stages of brain ischemia, transient SMC but not pericyte constrictions were a major cause of hypoperfusion leading to thrombosis and distal microvascular occlusions. Thus, capillary pericytes are not contractile, and regulation of cerebral blood flow in physiological and pathological conditions is mediated by arteriolar SMCs.


Assuntos
Isquemia Encefálica , Córtex Cerebral/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Contração Muscular/fisiologia , Miócitos de Músculo Liso/fisiologia , Pericitos/fisiologia , Actinas/metabolismo , Animais , Arteríolas/fisiologia , Encéfalo/irrigação sanguínea , Cálcio/metabolismo , Capilares/fisiologia , Humanos , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia
3.
Sci Transl Med ; 7(291): 291ra93, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26062845

RESUMO

The transcription factor nuclear factor κB (NFκB) is a central regulator of inflammation, and genome-wide association studies in subjects with autoimmune disease have identified a number of variants within the NFκB signaling cascade. In addition, causal variant fine-mapping has demonstrated that autoimmune disease susceptibility variants for multiple sclerosis (MS) and ulcerative colitis are strongly enriched within binding sites for NFκB. We report that MS-associated variants proximal to NFκB1 and in an intron of TNFRSF1A (TNFR1) are associated with increased NFκB signaling after tumor necrosis factor-α (TNFα) stimulation. Both variants result in increased degradation of inhibitor of NFκB α (IκBα), a negative regulator of NFκB, and nuclear translocation of p65 NFκB. The variant proximal to NFκB1 controls signaling responses by altering the expression of NFκB itself, with the GG risk genotype expressing 20-fold more p50 NFκB and diminished expression of the negative regulators of the NFκB pathway: TNFα-induced protein 3 (TNFAIP3), B cell leukemia 3 (BCL3), and cellular inhibitor of apoptosis 1 (CIAP1). Finally, naïve CD4 T cells from patients with MS express enhanced activation of p65 NFκB. These results demonstrate that genetic variants associated with risk of developing MS alter NFκB signaling pathways, resulting in enhanced NFκB activation and greater responsiveness to inflammatory stimuli. As such, this suggests that rapid genetic screening for variants associated with NFκB signaling may identify individuals amenable to NFκB or cytokine blockade.


Assuntos
Autoimunidade/genética , Predisposição Genética para Doença , Inflamação/genética , NF-kappa B/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Transdução de Sinais/genética , Fatores Etários , Alelos , Linfócitos T CD4-Positivos/imunologia , Estudos de Casos e Controles , Núcleo Celular/metabolismo , Citocinas/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/sangue , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Transporte Proteico , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fatores de Risco , Caracteres Sexuais , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
4.
Sci Transl Med ; 6(226): 226ra31, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24598589

RESUMO

Occlusion of the microvasculature by blood clots, atheromatous fragments, or circulating debris is a frequent phenomenon in most human organs. Emboli are cleared from the microvasculature by hemodynamic pressure and the fibrinolytic system. An alternative mechanism of clearance is angiophagy, in which emboli are engulfed by the endothelium and translocate through the microvascular wall. We report that endothelial lamellipodia surround emboli within hours of occlusion, markedly reducing hemodynamic washout and tissue plasminogen activator-mediated fibrinolysis in mice. Over the next few days, emboli are completely engulfed by the endothelium and extravasated into the perivascular space, leading to vessel recanalization and blood flow reestablishment. We find that this mechanism is not limited to the brain, as previously thought, but also occurs in the heart, retina, kidney, and lung. In the lung, emboli cross into the alveolar space where they are degraded by macrophages, whereas in the kidney, they enter the renal tubules, constituting potential routes for permanent removal of circulating debris. Retina photography and angiography in patients with embolic occlusions provide indirect evidence suggesting that angiophagy may also occur in humans. Thus, angiophagy appears to be a ubiquitous mechanism that could be a therapeutic target with broad implications in vascular occlusive disorders. Given its biphasic nature-initially causing embolus retention, and subsequently driving embolus extravasation-it is likely that different therapeutic strategies will be required during these distinct post-occlusion time windows.


Assuntos
Embolia/patologia , Fagocitose , Vasos Retinianos/patologia , Animais , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Circulação Coronária , Fibrina/química , Fibrinólise , Fundo de Olho , Proteínas de Fluorescência Verde/metabolismo , Hemodinâmica , Humanos , Túbulos Renais/irrigação sanguínea , Pulmão/irrigação sanguínea , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , Microcirculação , Microglia/metabolismo , Microscopia Eletrônica de Transmissão , Microvasos , Monócitos/citologia , Retina/metabolismo , Trombose
5.
PLoS One ; 8(6): e64684, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23750212

RESUMO

Stroke has a high incidence in the elderly. Stroke enters the chronic phase 3 months after initial stroke onset. Currently, there is no pharmaceutical treatment available for chronic stroke. We have demonstrated the therapeutic effects of the combination of stem cell factor (SCF) and granulocyte-colony stimulating factor (G-CSF) (SCF+G-CSF) on chronic stroke. However, it remains unclear how SCF+G-CSF repairs the brain in chronic stroke. In this study, we determined the effects of SCF+G-CSF on neuronal network remodeling in the aged brain of chronic stroke. Cortical brain ischemia was produced in 16-18 month-old transgenic mice expressing yellow fluorescent protein in layer V pyramidal neurons. SCF+G-CSF was subcutaneously injected for 7 days beginning at 3.5 months post-ischemia. Using both live brain imaging and immunohistochemistry, we observed that SCF+G-CSF increased the mushroom-type spines on the apical dendrites of layer V pyramidal neurons adjacent to the infarct cavities 2 and 6 weeks after treatment. SCF+G-CSF also augmented dendritic branches and post-synaptic density protein 95 puncta in the peri-infarct cortex 6 weeks after treatment. These data suggest that SCF+G-CSF treatment in chronic stroke remodels neural circuits in the aged brain. This study provides evidence to support the development of a new therapeutic strategy for chronic stroke.


Assuntos
Envelhecimento , Espinhas Dendríticas/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Rede Nervosa/efeitos dos fármacos , Fator de Células-Tronco/farmacologia , Acidente Vascular Cerebral/patologia , Animais , Doença Crônica , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Interações Medicamentosas , Guanilato Quinases/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Rede Nervosa/fisiopatologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Fatores de Tempo
6.
Recent Pat CNS Drug Discov ; 8(1): 2-12, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23173646

RESUMO

Stroke is a serious cerebrovascular disease that causes high mortality and persistent disability in adults worldwide. Stroke is also an enormous public health problem and a heavy public financial burden in the United States. Treatment for stroke is very limited. Thrombolytic therapy by tissue plasminogen activator (tPA) is the only approved treatment for acute stroke, and no effective treatment is available for chronic stroke. Developing new therapeutic strategies, therefore, is a critical need for stroke treatment. This article summarizes the discovery of new routes of treatment for acute and chronic stroke using two hematopoietic growth factors, stem cell factor (SCF) and granulocyte-colony stimulating factor (G-CSF). In a study of acute stroke, SCF and G-CSF alone or in combination displays neuroprotective effects in an animal model of stroke. SCF appears to be the optimal treatment for acute stroke as the functional outcome is superior to G-CSF alone or in combination (SCF+G-CSF); however, SCF+G-CSF does show better functional recovery than G-CSF. In a chronic stroke study, the therapeutic effects of SCF and G-CSF alone or in combination appear differently as compared with their effects on the acute stroke. SCF+G-CSF induces stable and long-lasting functional improvement; SCF alone also improves functional outcome but its effectiveness is less than SCF+G-CSF, whereas G-CSF shows no therapeutic effects. Although the mechanism by which SCF+G-CSF repairs the brain in chronic stroke remains poorly understood, our recent findings suggest that the SCF+G-CSF-induced functional improvement in chronic stroke is associated with a contribution to increasing angiogenesis and neurogenesis through bone marrow-derived cells and the direct effects on stimulating neurons to form new neuronal networks. These findings would assist in developing new treatment for stroke. The article presents some promising patents on role of stem cell factor and granulocyte-colony stimulating factor in treatment of stroke.


Assuntos
Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Indutores da Angiogênese/uso terapêutico , Animais , Doença Crônica , Quimioterapia Combinada , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Neurogênese/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Fator de Células-Tronco/administração & dosagem
7.
Mol Cell Neurosci ; 46(1): 325-32, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21040786

RESUMO

The cytokine TWEAK is expressed in the brain and is induced in cerebral ischemia and other brain disorders. TWEAK regulates proliferation and differentiation of progenitor cells but its effect on adult neural progenitor cells is still unknown. Therefore, we investigated the proliferation of neural progenitor cells from the subventricular zone of adult mice in response to TWEAK treatment. TWEAK inhibited proliferation of neural progenitor cells through its membrane receptor Fn14. The reduced proliferation was not due to cell death. By using a reporter assay we found that TWEAK activated the transcription factor NF-κB in adult neural progenitor cells. Blockade of NF-κB signaling reversed the inhibition of cell proliferation by TWEAK. In addition, TWEAK induced neuronal differentiation of neural progenitor cells and lowered the expression of hes1, a transcription factor that prevents neuronal differentiation. In adult mice deficient of the TWEAK receptor Fn14, neurogenesis was reduced in the subventricular zone. In conclusion, our data show that TWEAK regulates adult neurogenesis in the subventricular zone by binding to the membrane receptor Fn14 and activating NF-κB.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Fatores de Necrose Tumoral/metabolismo , Animais , Citocina TWEAK , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor de TWEAK , Fatores de Necrose Tumoral/genética
8.
FASEB J ; 24(3): 788-98, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19884325

RESUMO

Activation of the cannabinoid 2 receptor (CB(2)) reduces ischemic injury in several organs. However, the mechanisms underlying this protective action are unclear. In a mouse model of ischemic stroke, we show that the CB(2) agonist JWH-133 (1 mg . kg(-1) . d(-1)) decreases the infarct size measured 3 d after onset of ischemia. The neuroprotective effect of JWH-133 was lost in CB(2)-deficient mice, confirming the specificity of JWH-133. Analysis of bone marrow chimeric mice revealed that bone marrow-derived cells mediate the CB(2) effect on ischemic brain injury. CB(2) activation reduced the number of neutrophils in the ischemic brain as shown by FACS analysis and by measuring the levels of the neutrophil marker enzyme myeloperoxidase. Indeed, we found in vitro that CB(2) activation inhibits adherence of neutrophils to brain endothelial cells. JWH-133 (1 microM) also interfered with the migration of neutrophils induced by the endogenous chemokine CXCL2 (30 ng/ml) through activation of the MAP kinase p38. This effect on neutrophils is likely responsible for the neuroprotection mediated by JWH-133 because JWH-133 was no longer protective when neutrophils were depleted. In conclusion, our data demonstrate that by activating p38 in neutrophils, CB(2) agonists inhibit neutrophil recruitment to the brain and protect against ischemic brain injury.-Murikinati, S., Jüttler, E., Keinert, T., Ridder, D. A., Muhammad, S., Waibler, Z., Ledent, C., Zimmer, A., Kalinke, U., Schwaninger, M. Activation of cannabinoid 2 receptors protects against cerebral ischemia by inhibiting neutrophil recruitment.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Neutrófilos/citologia , Receptor CB2 de Canabinoide/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Encéfalo/citologia , Canabinoides/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL2/metabolismo , Células Endoteliais/citologia , Citometria de Fluxo , Immunoblotting , Interleucina-1beta/metabolismo , Masculino , Camundongos , Neutrófilos/efeitos dos fármacos , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/metabolismo
9.
J Neurosci Methods ; 184(1): 95-103, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19660497

RESUMO

Reducing post-stroke disability is the major goal of stroke therapy. Consequently, functional testing is essential in experimental stroke studies to increase the predictive value of animal models. We used several sensory and motor tests to assess functional disability in a mouse model of permanent distal middle cerebral artery occlusion (pdMCAO) that induced mainly cortical infarcts. Gait dynamics were transiently disturbed after pdMCAO as measured by different analysis techniques. Stance and brake duration were shorter after pdMCAO. Consistent with sensory and motor deficits the latency to move was prolonged up to 14 days after pdMCAO and the performance in the corner test and handedness were affected on day 1 or 2 after pdMCAO. Heart rate was decreased and heart rate variability were increased after pdMCAO indicating sympathetic-parasympathetic imbalance. In summary, pdMCAO-induced cortical infarcts lead to clinically relevant sensory, motor and cardiac autonomic dysfunction in mice. The present study provides a basis to explore the potential of functional testing for neuroprotection and neuroregeneration after stroke.


Assuntos
Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/fisiopatologia , Animais , Peso Corporal , Encéfalo/patologia , Infarto Encefálico/patologia , Infarto Encefálico/fisiopatologia , Avaliação da Deficiência , Eletrocardiografia , Lateralidade Funcional , Marcha , Frequência Cardíaca , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Transtornos dos Movimentos/patologia , Transtornos dos Movimentos/fisiopatologia , Postura , Fatores de Tempo
10.
J Neurosci ; 28(46): 12023-12031, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19005067

RESUMO

In ischemic stroke, the necrotic core is surrounded by a zone of inflammation, in which delayed cell death aggravates the initial insult. Here, we provide evidence that the receptor for advanced glycation end products (RAGE) functions as a sensor of necrotic cell death and contributes to inflammation and ischemic brain damage. The RAGE ligand high mobility group box 1 (HMGB1) was elevated in serum of stroke patients and was released from ischemic brain tissue in a mouse model of cerebral ischemia. A neutralizing anti-HMGB1 antibody and HMGB1 box A, an antagonist of HMGB1 at the receptor RAGE, ameliorated ischemic brain damage. Interestingly, genetic RAGE deficiency and the decoy receptor soluble RAGE reduced the infarct size. In vitro, expression of RAGE in (micro)glial cells mediated the toxic effect of HMGB1. Addition of macrophages to neural cultures further enhanced the toxic effect of HMGB1. To test whether immigrant macrophages in the ischemic brain mediate the RAGE effect, we generated chimeric mice by transplanting RAGE(-/-) bone marrow to wild-type mice. RAGE deficiency in bone marrow-derived cells significantly reduced the infarct size. Thus, HMGB1-RAGE signaling links necrosis with macrophage activation and may provide a target for anti-inflammatory therapy in stroke.


Assuntos
Infarto Encefálico/metabolismo , Isquemia Encefálica/metabolismo , Encefalite/metabolismo , Proteína HMGB1/metabolismo , Receptores Imunológicos/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos/farmacologia , Transplante de Medula Óssea/métodos , Infarto Encefálico/genética , Infarto Encefálico/fisiopatologia , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatologia , Morte Celular/genética , Células Cultivadas , Encefalite/genética , Encefalite/fisiopatologia , Feminino , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/genética , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , Necrose/genética , Necrose/metabolismo , Necrose/fisiopatologia , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quimeras de Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...