Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Yao Xue Xue Bao ; 49(10): 1413-7, 2014 Oct.
Artigo em Zh | MEDLINE | ID: mdl-25577871

RESUMO

The aim of this study is to explore the tissue distribution of PEGylated puerarin in acute myocardial ischemia model rats. Healthy male SD rats were randomly divided into two groups (30 each). Both were given PEGylated puerarin at a dose of 488 mg x kg(-1). After 5 min of medication, one group was normal rats, another group with acute myocardial ischemia was established by peritoneal injection of 50 mg x kg(-1) isoprenaline. After administration, the animals were executed at 30, 60, 90, 120, 150 and 180 min, then heart, liver, spleen, lung, kidney were extracted. The content of puerarin in organ tissue was determined by HPLC. The results showed that the AUC of tissue distribution of PEGylated puerarin in normal rats was liver > kidney > heart ≈ spleen > lung > brain. While the AUC of tissue distribution of PEGylated puerarin in acute myocardial ischemia model rats was liver ≈ heart > kidney > lung ≈ spleen > brain. AUC(heart) of PEGylated puerarin in acute myocardial ischemia model rats was 1.7 times than that of the normal rats, and there was significant difference (P < 0.05). Thus, PEGylated puerarin had a good heart-targeting property in early myocardial infarction area, drugs could accumulate in the ischemic myocardium. It provided important information for further study and clinic use of PEGylated puerarin.


Assuntos
Isoflavonas/farmacocinética , Isquemia Miocárdica/metabolismo , Animais , Encéfalo/metabolismo , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Baço/metabolismo , Distribuição Tecidual
2.
Nat Med ; 30(6): 1680-1688, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38740994

RESUMO

Emotional distress (ED), commonly characterized by symptoms of depression and/or anxiety, is prevalent in patients with cancer. Preclinical studies suggest that ED can impair antitumor immune responses, but few clinical studies have explored its relationship with response to immune checkpoint inhibitors (ICIs). Here we report results from cohort 1 of the prospective observational STRESS-LUNG study, which investigated the association between ED and clinical efficacy of first-line treatment of ICIs in patients with advanced non-small-cell lung cancer. ED was assessed by Patient Health Questionnaire-9 and Generalized Anxiety Disorder 7-item scale. The study included 227 patients with 111 (48.9%) exhibiting ED who presented depression (Patient Health Questionnaire-9 score ≥5) and/or anxiety (Generalized Anxiety Disorder 7-item score ≥5) symptoms at baseline. On the primary endpoint analysis, patients with baseline ED exhibited a significantly shorter median progression-free survival compared with those without ED (7.9 months versus 15.5 months, hazard ratio 1.73, 95% confidence interval 1.23 to 2.43, P = 0.002). On the secondary endpoint analysis, ED was associated with lower objective response rate (46.8% versus 62.1%, odds ratio 0.54, P = 0.022), reduced 2-year overall survival rate of 46.5% versus 64.9% (hazard ratio for death 1.82, 95% confidence interval 1.12 to 2.97, P = 0.016) and detriments in quality of life. The exploratory analysis indicated that the ED group showed elevated blood cortisol levels, which was associated with adverse survival outcomes. This study suggests that there is an association between ED and worse clinical outcomes in patients with advanced non-small-cell lung cancer treated with ICIs, highlighting the potential significance of addressing ED in cancer management. ClinicalTrials.gov registration: NCT05477979 .


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Inibidores de Checkpoint Imunológico , Neoplasias Pulmonares , Angústia Psicológica , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/efeitos adversos , Feminino , Masculino , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Pessoa de Meia-Idade , Idoso , Estudos Prospectivos , Depressão/tratamento farmacológico , Ansiedade/tratamento farmacológico , Resultado do Tratamento , Intervalo Livre de Progressão , Adulto , Idoso de 80 Anos ou mais
3.
Alzheimers Dement ; 9(6): 666-76, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23411393

RESUMO

OBJECTIVE: We wished to develop a highly selective positron emission tomography (PET) imaging agent targeting PHF-tau in human Alzheimer's disease (AD) brains. METHODS: To screen potential tau binders, human AD brain sections were used as a source of native paired helical filament (PHF)-tau and Aß rather than synthetic tau aggregates or Aß fibrils generated in vitro to measure the affinity and selectivity of [(18)F]T807 to tau and Aß. Brain uptake and biodistribution of [(18)F]T807 in mice were also tested. RESULTS: In vitro autoradiography results show that [(18)F]T807 exhibits strong binding to PHF-tau-positive human brain sections. A dissociation constant (Kd) of [(18)F]T807 (14.6 nM) was measured using brain sections from the frontal lobe of AD patients. A comparison of autoradiography and double immunohistochemical staining of PHF-tau and Aß on adjacent sections demonstrated that [(18)F]T807 binding colocalized with immunoreactive PHF-tau pathology, but did not highlight Aß plaques. In vivo studies in mice demonstrated that [(18)F]T807 was able to cross the blood-brain barrier and washed out quickly. CONCLUSIONS: [(18)F]T807 demonstrates high affinity and selectivity to PHF-tau as well as favorable in vivo properties, making this a promising candidate as an imaging agent for AD.


Assuntos
Doença de Alzheimer/diagnóstico , Encéfalo/diagnóstico por imagem , Radioisótopos de Flúor , Proteínas tau/química , Proteínas tau/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/metabolismo , Animais , Autorradiografia , Encéfalo/patologia , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Distribuição Tecidual , Proteínas tau/genética
4.
J Int Med Res ; 49(4): 3000605211004021, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33794676

RESUMO

OBJECTIVE: This study aimed to identify a predictive marker of response to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in patients with EGFR-mutant advanced lung adenocarcinoma. METHODS: A cohort of 190 patients with EGFR-mutant advanced lung adenocarcinoma was analyzed. Receiver operating characteristic curve analysis was used to evaluate the optimal cutoffs for fibrinogen levels, the neutrophil-to-lymphocyte ratio (NLR), and the platelet-to-lymphocyte ratio (PLR) for predicting progression-free survival (PFS). Univariate and multivariate survival analyses were performed to identify factors correlated with PFS and overall survival (OS). RESULTS: High NLR was associated with worse performance status. In univariate analysis, fibrinogen levels, NLR, and PLR were correlated with OS and PFS. In multivariate analysis, all three variables remained predictive of OS, whereas only fibrinogen levels and PLR were independent prognostic factors for PFS. Furthermore, the combination of fibrinogen levels and PLR (F-PLR score) could stratify patients into three groups with significantly different prognoses, and the score was independently predictive of survival. CONCLUSION: The F-PLR score predicted the prognosis of patients with EGFR-mutant advanced lung adenocarcinoma who received EGFR-TKIs, and this score may serve as a convenient blood-based marker for identifying high-risk patients.


Assuntos
Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Receptores ErbB/genética , Fibrinogênio , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Linfócitos , Neutrófilos , Prognóstico , Inibidores de Proteínas Quinases/uso terapêutico , Estudos Retrospectivos
5.
Biol Chem ; 391(4): 345-55, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20180644

RESUMO

Tissue kallikrein (KLK1) processes low-molecular weight kininogen to produce vasoactive kinins, which exert biological functions via kinin receptor signaling. Using various delivery approaches, we have demonstrated that tissue kallikrein through kinin B2 receptor signaling exhibits a wide spectrum of beneficial effects by reducing cardiac and renal injuries, restenosis and ischemic stroke, and by promoting angiogenesis and skin wound healing, independent of blood pressure reduction. Protection by tissue kallikrein in oxidative organ damage is attributed to the inhibition of apoptosis, inflammation, hypertrophy and fibrosis. Tissue kallikrein also enhances neovascularization in ischemic heart and limb. Moreover, tissue kallikrein/kinin infusion not only prevents but also reverses kidney injury, inflammation and fibrosis in salt-induced hypertensive rats. Furthermore, there is a wide time window for kallikrein administration in protection against ischemic brain infarction, as delayed kallikrein infusion for 24 h after cerebral ischemia in rats is effective in reducing neurological deficits, infarct size, apoptosis and inflammation. Importantly, in the clinical setting, human tissue kallikrein has been proven to be effective in the treatment of patients with acute brain infarction when injected within 48 h after stroke onset. Finally, kallikrein promotes skin wound healing and keratinocyte migration by direct activation of protease-activated receptor 1.


Assuntos
Doenças Cardiovasculares/metabolismo , Transtornos Cerebrovasculares/metabolismo , Nefropatias/metabolismo , Pele/lesões , Pele/metabolismo , Calicreínas Teciduais/metabolismo , Cicatrização , Animais , Doenças Cardiovasculares/fisiopatologia , Doenças Cardiovasculares/prevenção & controle , Transtornos Cerebrovasculares/fisiopatologia , Transtornos Cerebrovasculares/prevenção & controle , Humanos , Nefropatias/prevenção & controle , Pele/patologia , Calicreínas Teciduais/antagonistas & inibidores , Calicreínas Teciduais/química , Calicreínas Teciduais/uso terapêutico
6.
J Oncol ; 2019: 5062394, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31031811

RESUMO

The synthetic house-tree-person (S-HTP) drawing test is a projective measure primarily designed to assess specific complex personality traits. It is widely used in general psychological problems and mental illness such as psychological crisis intervention. Applicability and validity of S-HTP drawing test in cancer patients suffering from anxiety are still unclear and there are no reports on such research. The aim of this study was to explore the prevalence of anxiety in cancer patients and to investigate the applicability of S-HTP drawing test in such patients. Self-rating anxiety scale (SAS) and the S-HTP drawing test were applied to 167 cancer patients (58.7% male; 41.3% female), 52.92±10.43 years old. On SAS, anxiety rate was found in 16.17% cancer patients. Using the evaluation results from SAS as the dependent variable and the anxiety drawing characteristics as the independent variables, the logistic regression equation was established, and 9 drawing features were employed in the regression equation (χ2=56.982, P≤0.001, Nagelkerke R2=0.492). It is concluded that there is a positive correlation between S-HTP drawing test and SAS for anxiety state of cancer patients (p<0.01). S-HTP drawing test and SAS have interrater reliability and test-retest reliability. Our findings indicate that the S-HTP drawing test could help in screening anxiety in cancer patients.

7.
J Nucl Med ; 60(8): 1154-1159, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30733317

RESUMO

The P2X7 receptor is an adenosine triphosphate-gated ion channel, which is abundantly expressed in glial cells within the central nervous system and in the periphery. P2X7 receptor activation leads to the release of the proinflammatory cytokine IL-1ß in the brain, and antagonism of the P2X7 receptor is a novel therapeutic strategy to dampen adenosine triphosphate-dependent IL-1ß signaling. PET ligands for the P2X7 receptor will not only be valuable to assess central target engagement of drug candidates but also hold promise as surrogate markers of central neuroinflammation. Herein we describe the in vitro and in vivo evaluation of 18F-JNJ-64413739, an 18F-labeled PET ligand for imaging the P2X7 receptor in the brain. Methods: P2X7 receptor affinity and specificity, pharmacokinetics, metabolic stability, blood-brain barrier permeability, and off-target binding of JNJ-64413739 were evaluated in a series of in vitro, ex vivo, and in vivo assays. 18F-JNJ-64413739 was radiolabeled via a one-step nucleophilic aromatic substitution. The tracer was also studied in rhesus macaques, and PET images were analyzed with an arterial plasma input function-based Logan graphical analysis. Results: The potency (half-maximal inhibitory concentration) of the P2X7 receptor antagonist JNJ-64413739 is 1.0 ± 0.2 nM and 2.0 ± 0.6 nM at the recombinant human and rat P2X7 receptor, respectively, and the binding affinity is 2.7 nM (rat cortex binding assay) and 15.9 nM (human P2X7 receptor). In nonhuman primate PET imaging studies, dose-dependent receptor occupancy of JNJ-54175446 was observed in 2 rhesus monkeys. At a 0.1 mg/kg dose (intravenous) of JNJ-54175446, the receptor occupancy was calculated to be 17% by Logan graphical analysis, whereas a dose of 2.5 mg/kg yielded a receptor occupancy of 60%. Conclusion: The preclinical evaluation of 18F-JNJ-64413739 demonstrates that the tracer engages the P2X7 receptor. Reproducible and dose-dependent receptor occupancy studies with the P2X7 receptor antagonist JNJ-54175446 were obtained in rhesus monkeys. This novel PET tracer exhibits in vitro and in vivo characteristics suitable for imaging the P2X7 receptor in the brain and warrants further studies in humans.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Peptídeos/farmacologia , Pirimidinas/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Feminino , Humanos , Técnicas In Vitro , Inflamação/diagnóstico por imagem , Cinética , Ligantes , Macaca mulatta , Masculino , Camundongos Knockout , Permeabilidade/efeitos dos fármacos , Tomografia por Emissão de Pósitrons , Ligação Proteica , Radioquímica , Ratos , Distribuição Tecidual , Resultado do Tratamento
8.
Mol Imaging Biol ; 21(5): 871-878, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30632003

RESUMO

PURPOSE: The P2X7 receptor, an adenosine triphosphate (ATP)-gated purinoreceptor, has emerged as one of the key players in neuroinflammatory processes. Therefore, developing a positron emission tomography (PET) tracer for imaging of P2X7 receptors in vivo presents a promising approach to diagnose, monitor, and study neuroinflammation in a variety of brain disorders. To fulfill the goal of developing a P2X7 PET ligand as a biomarker of neuroinflammation, [18F]JNJ-64413739 has been recently disclosed. PROCEDURES: We evaluated [18F]JNJ-64413739 in a rat model of neuroinflammation induced by an intracerebral injection of lipopolysaccharide (LPS). In vivo brain uptake was determined by PET imaging. Upregulation of neuroinflammatory biomarkers was determined by quantitative polymerase chain reaction (qPCR). Distribution of the tracer in the brain was determined by ex vivo autoradiography (ARG). The specificity of [18F]JNJ-64413739 was confirmed by performing blocking experiments with the P2X7 antagonist JNJ-54175446. RESULTS: Brain regions of rats injected with LPS had a significantly increased uptake (34 % ± 3 % s.e.m., p = 0.036, t test, standardized uptake value measured over the entire scanning period) of [18F]JNJ-64413739 relative to the corresponding brain regions of control animals injected with phosphate-buffered saline (PBS). The uptake in the contralateral regions and cerebellum was not significantly different between the groups of animals. The increase in uptake of [18F]JNJ-64413739 at the LPS-injected site observed by PET imaging was concordant with ex vivo ARG, upregulation of neuroinflammatory biomarkers, and elevated P2X7 expression levels. CONCLUSIONS: While further work is needed to study [18F]JNJ-64413739 in other types of neuroinflammation, the current results favorably characterize [18F]JNJ-64413739 as a potential PET tracer of central neuroinflammation.


Assuntos
Encéfalo/diagnóstico por imagem , Inflamação/diagnóstico por imagem , Inflamação/patologia , Peptídeos/química , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Receptores Purinérgicos P2X7/metabolismo , Animais , Biomarcadores/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Lipopolissacarídeos , Peptídeos/farmacocinética , Projetos Piloto , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Piridinas/química , Ratos , Triazóis/química
9.
J Med Chem ; 62(6): 2974-2987, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30810314

RESUMO

In Alzheimer's disease, the density and spread of aggregated tau protein track well with neurodegeneration and cognitive decline, making the imaging of aggregated tau a compelling biomarker. A structure-activity relationship exploration around an isoquinoline hit, followed by an exploration of tolerated fluorination positions, allowed us to identify 9 (JNJ-64326067), a potent and selective binder to aggregated tau with a favorable pharmacokinetic profile and no apparent off-target binding. This was confirmed in rat and monkey positron emission tomography studies using [18F]9.


Assuntos
Descoberta de Drogas , Radioisótopos de Flúor/metabolismo , Isoquinolinas/farmacocinética , Tomografia por Emissão de Pósitrons , Piridinas/farmacocinética , Proteínas tau/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Feminino , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Hepatócitos/metabolismo , Humanos , Isoquinolinas/química , Macaca mulatta , Masculino , Piridinas/química , Ratos , Ratos Wistar , Relação Estrutura-Atividade
10.
Biotechnol Bioeng ; 99(2): 475-84, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17680664

RESUMO

Mucopolysaccharidosis Type I, Hurler's Syndrome, is a lysosomal storage disorder that affects the brain. The missing enzyme, alpha-L-iduronidase (IDUA), does not cross the blood-brain barrier (BBB). To enable BBB transport of the enzyme, human IDUA was fused to the carboxyl terminus of the heavy chain of a chimeric monoclonal antibody (MAb) to the human insulin receptor (HIR). The HIRMAb crosses the BBB on the endogenous insulin receptor, and acts as a molecular Trojan horse to ferry into brain the IDUA. Transfection of COS cells resulted in high levels of IDUA enzyme activity both in the medium and in the intracellular space. The size of the fusion heavy chain, as measured with Western blotting and antibodies to either human IDUA or human IgG, was increased about 80 kDa, relative to the size of the heavy chain of the parent HIRMAb. The IDUA enzyme specific activity of the affinity purified HIRMAb-IDUA fusion protein was 363 +/- 37 U/microg protein, which is comparable to specific activity of recombinant IDUA. The accumulation of glycosoaminoglycans in Hurler fibroblasts was decreased 70% by treatment with the HIRMAb-IDUA fusion protein. Confocal microscopy showed targeting of the fusion protein to the lysosome. The HIRMAb-IDUA fusion protein bound with high affinity to the HIR, and was rapidly transported into the brain of the adult Rhesus monkey following intravenous administration. The HIRMAb-IDUA fusion protein is a new treatment for Hurler's syndrome, which has been specifically engineered to cross the human BBB.


Assuntos
Anticorpos Monoclonais/farmacocinética , Barreira Hematoencefálica/fisiologia , Iduronidase/farmacocinética , Mucopolissacaridose I/terapia , Engenharia de Proteínas/métodos , Receptor de Insulina/química , Proteínas Recombinantes de Fusão/farmacocinética , Animais , Anticorpos Monoclonais/administração & dosagem , Barreira Hematoencefálica/efeitos dos fármacos , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Fibroblastos , Humanos , Iduronidase/administração & dosagem , Iduronidase/química , Mucopolissacaridose I/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/química , Transfecção/métodos
11.
Clin Hemorheol Microcirc ; 65(1): 11-22, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27258204

RESUMO

OBJECTIVE: This study aimed to examine the influence of oral acetylsalicylic acid on blood fluidity and infusion speed in the cancer patients with Peripherally Inserted Central Catheter (PICC). BACKGROUND: PICC is placed for prolonged chemotherapy of cancer patients. The fibrin sheaths, which consist of cellular substance and non-cellular substance, generate at the place of insertion and grow down all over the catheter. Finally they cover the vent of the catheter and lead to catheter dysfunctions such as the decrease of infusion speed. In addition, the high viscosity status of cancer patients could lead to acute embolization, which adds to the high risk of death. DESIGN: Randomized controlled trial. METHODS: This research was carried out between April 2013 and January 2014 in the second hospital of Xiangya, Central South University in Changsha, China. Initially 96 cancer participants with PICC were chosen and randomly allocated to experimental and control group. The participants of the experimental group were conducted route PICC maintain technique and took acetylsalicylic acid 100 mg per day after dinner, while the control group received route PICC maintain technique only. The infusion speed and hemorheology indexes of the two groups were tested before our study and at the end of the 2nd and 4th months with several instruments. RESULTS: Repeated measures analysis of variance indicated that taking acetylsalicylic acid orally had significant main effect on high shear blood viscosity and red blood cell deformability index (P < 0.05), and it also had significant main effect as well as time effect on plasma viscosity (P < 0.05); and time had significant main effect as well as interaction effect with oral acetylsalicylic acid on low shear blood viscosity and red blood cell aggregation index (P < 0.05). Repeated measures ANOVA also showed that taking acetylsalicylic acid orally had significant main effect, time effect and interaction effect on infusion speed (P < 0.01). CONCLUSION: Oral acetylsalicylic acid could improve hemorheology condition and the infusion speed related to fibrin sheath in the cancer patients.


Assuntos
Aspirina/uso terapêutico , Cateterismo Periférico/instrumentação , Neoplasias/terapia , Administração Oral , Cateterismo Periférico/métodos , Feminino , Hemorreologia , Humanos , Masculino , Pessoa de Meia-Idade
12.
Hum Gene Ther ; 17(2): 206-19, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16454654

RESUMO

Stroke-induced neurological deficits and mortality are often associated with timing of treatment after the onset of stroke. We showed that local delivery of the human tissue kallikrein gene into rat brain immediately after middle cerebral artery occlusion (MCAO) exerts neuroprotection. In this study, we investigated the effect of systemic delivery of the kallikrein gene 8 hr after MCAO. Expression of recombinant human tissue kallikrein after gene transfer was identified in the ischemic brain region and blood vessels. Intravenous injection of adenovirus encoding the kallikrein gene significantly reduced neurological deficit scores 2 and 7 days after gene transfer. Kallikrein gene transfer also reduced ischemia-reperfusion (I/R)-induced cerebral infarction and promoted the survival and migration of glial cells from penumbra to the ischemic core from 3 to 14 days after gene delivery. Kallikrein reduced I/R-induced apoptosis of neuronal cells and inhibited inflammatory cell accumulation in the ischemic brain. These effects were blocked by the kinin B2 receptor antagonist icatibant. In addition, kallikrein enhanced angiogenesis and promoted neurogenesis after I/R and the stimulatory effect of kinin on neuronal cell proliferation was confirmed in primary cultured neuronal cells. The protective effects of kallikrein, through the kinin B2 receptor, were accompanied by increased cerebral nitric oxide and Bcl-2 levels, Akt phosphorylation, and reduced NAD(P)H oxidase activity, superoxide production, Bax levels, and caspase-3 activity. These results indicate that delayed systemic administration of the kallikrein gene after onset of stroke protects against ischemic brain injury by inhibiting apoptosis and inflammation and by promoting angiogenesis and neurogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Inflamação/tratamento farmacológico , Calicreínas/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/prevenção & controle , Animais , Apoptose/genética , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Células Cultivadas , Técnicas de Transferência de Genes , Humanos , Calicreínas/genética , Masculino , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/metabolismo , Proteína Oncogênica v-akt/efeitos dos fármacos , Proteína Oncogênica v-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Traumatismo por Reperfusão/terapia
13.
Life Sci ; 76(21): 2457-71, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15763077

RESUMO

Nitric oxide (NO) has been shown to play a key role in the regulation of cardiac hypertrophy and fibrosis in response to myocardial ischemia in part by antagonizing the action of angiotensin II (Ang II). In this study, we investigated the potential protective role of human endothelial nitric oxide synthase (eNOS) in left ventricular (LV) remodeling after myocardial infarction (MI) by a somatic gene transfer approach. Male Wistar rats underwent coronary artery ligation to induce MI. One week after surgery, adenovirus encoding the human eNOS or luciferase gene under the control of the CMV promoter/enhancer was injected into rats via the tail vein, and animals were sacrificed at 1 and 5 weeks after gene transfer. Successful gene transfer was evaluated based on increased levels of NO and cGMP in the heart, measured at one week after eNOS gene delivery. Six weeks after MI, the LV end-diastolic pressure, heart weight, LV axis length and cardiomyocyte size were markedly increased compared to the Sham group, while eNOS gene delivery significantly reduced these parameters. Rats receiving control virus developed considerably more fibrotic lesions identified by Sirius Red staining and collagen I immunostaining compared to Sham rats, and eNOS gene delivery significantly reduced collagen accumulation. eNOS gene transfer also reduced TUNEL-positive apoptotic cells. The cardioprotective effect of NO was accompanied by reduced NADH and NADPH oxidase activities and superoxide formation, TGF-beta1 and p27 levels, JNK activation, NF-kappa B nuclear translocation, and caspase-3 activity. This study shows that NO may play an important role in attenuating cardiac remodeling and apoptosis after myocardial infarction via suppression of oxidative stress-mediated signaling pathways.


Assuntos
Terapia Genética/métodos , Infarto do Miocárdio/terapia , Óxido Nítrico Sintase/genética , Estresse Oxidativo/genética , Remodelação Ventricular/genética , Adenoviridae , Animais , Apoptose/fisiologia , Compostos Azo , Caspase 3 , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Colágeno , Vasos Coronários/cirurgia , GMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Vetores Genéticos , Ventrículos do Coração/patologia , Humanos , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ligadura , Luciferases/genética , MAP Quinase Quinase 4 , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NAD/metabolismo , NADP/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III , Tamanho do Órgão , Ratos , Ratos Wistar , Superóxidos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas Supressoras de Tumor/metabolismo , Função Ventricular
14.
Arch Gerontol Geriatr ; 61(1): 72-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25957866

RESUMO

BACKGROUND: The present study aimed to explore the association between RDW and CAS in patients with ischemic stroke, expecting to find a new and significant diagnosis index for clinical practice. METHODS: This cross-sectional study involves 432 consecutive patients with primary ischemic stroke (within 72 h). All subjects were confirmed by magnetic resonance imaging, and underwent physical examination, laboratory tests and carotid ultrasonography check. Finally, 392 patients were included according to the exclusion criteria. The odds ratios of independent variables were calculated using stepwise multiple logistic regression. RESULTS: Carotid intimal-medial thickness (IMT) and RDW are both significantly different between CAS group and control group. Univariate analyses show that high-sensitive C-reactive protein (Hs-CRP) and RDW (r=0.436) are both in significantly positive association with IMT. Stepwise multiple logistic regression shows that RDW is an independent protective factor of CAS in patients with ischemic stroke. Compared with the lowest quartile, the second to fourth quartiles are 1.13 (95% CI: 1.13-3.05), 2.02 (95% CI: 1.66-4.67), and 3.10 (95% CI: 2.46-7.65), respectively. CONCLUSION: The present study suggested that RDW level were higher than non-CAS in patients with primary ischemic stroke. Our results facilitated a bridge to connect RDW with ischemic stroke and further confirmed the role of RDW in the progression of the ischemic stroke.


Assuntos
Isquemia Encefálica/sangue , Doenças das Artérias Carótidas/sangue , Idoso , Isquemia Encefálica/epidemiologia , Isquemia Encefálica/etiologia , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/complicações , Doenças das Artérias Carótidas/diagnóstico , China/epidemiologia , Estudos Transversais , Índices de Eritrócitos , Feminino , Humanos , Incidência , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Razão de Chances , Fatores de Risco
16.
Hum Gene Ther ; 15(12): 1243-54, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15684700

RESUMO

Adrenomedullin (AM) has been shown to protect against ischemia/reperfusion-induced myocardial infarction and apoptosis. In the present study, we examined the potential neuroprotective action of delayed AM gene transfer in cerebral ischemia. Three days after a 1-hr occlusion of the middle cerebral artery (MCAO), rats were injected intravenously with adenovirus harboring human AM cDNA. The experiment was terminated 7 days after MCAO. AM gene transfer significantly reduced cerebral infarct size compared with that of rats before virus injection and compared with that of rats injected with control virus. The expression of recombinant human AM was identified in ischemic brain by immunostaining. Morphological analyses showed that AM gene transfer enhanced the survival and migration of astrocytes into the ischemic core. Cerebral ischemia markedly increased astrocyte apoptosis, and AM gene delivery significantly reduced apoptosis to near normal levels as seen in sham control rats. Similarly, in primary cultured astrocytes, AM stimulated cell migration and inhibited hypoxia/reoxygenation-induced apoptosis. The effects of AM on both migration and apoptosis were abolished by calcitonin gene-related peptide [CGRP(8-37)], an AM receptor antagonist. Enhanced cell survival after AM gene transfer was accompanied by markedly increased cerebral nitric oxide and Bcl-2 levels, as well as Akt and GSK-3beta phosphorylation, but reduced NADPH oxidase activity and superoxide production. Inactivation of GSK-3beta by phosphorylation led to reduced GSK-3beta activity and caspase- 3 activation. These results indicate that exogenous AM provides neuroprotection against cerebral ischemia injury by enhancing astrocyte survival and migration and inhibiting apoptosis through suppression of oxidative stress-mediated signaling events.


Assuntos
Astrócitos/citologia , Isquemia Encefálica/prevenção & controle , Movimento Celular/genética , Técnicas de Transferência de Genes , Fármacos Neuroprotetores , Peptídeos/genética , Adrenomedulina , Animais , Apoptose/genética , Astrócitos/fisiologia , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Caspases/metabolismo , Sobrevivência Celular , Células Cultivadas , Infarto Cerebral/patologia , Infarto Cerebral/prevenção & controle , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Imuno-Histoquímica , Masculino , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-bcl-2 , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo
17.
Chin Med J (Engl) ; 115(4): 494-7, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12133282

RESUMO

OBJECTIVE: To study the effects of focal adhesion kinase (FAK) phosphorylation on smooth muscle cells (SMCs) adhesion and migration stimulated by fibronectin. METHODS: Adhesion and migration of cultured SMCs were stimulated by different concentrations of fibronectin (FN), FAK and its phosphorylation were detected by immunoprecipitation and Western blot. FAK antisense oligodeoxynucleotides (ODNs) were transfected into SMCs by cationic lipid to investigate its modulatory effects on tyrosine phosphorylation. SMCs adhesion and migration were also measured by morphological enumeration and modified Boyden Chambers, respectively. RESULTS: FAK were expressed when SMCs adhesion and migration were successfully simulated by different concentrations of FN. FAK phosphorylation were detected only at 20 microg/ml FN or more. FAK antisense ODNs were transfected efficiently by cationic lipid and FAK phosphorylation was inhibited substantially. The SMCs migration rate in the 5 - 60 microg/ml FN groups was reduced by 17.89% - 27.67%. Cell migration stimulated by FN at 10, 20, 40 and 60 microg/ml were reduced by 23.26%, 21.63%, 19.31% and 17.88%, respectively (P < 0.05). CONCLUSIONS: FAK phosphorylation and FAK-mediated signal transduction play important roles in SMCs adhesion and migration stimulated by ECM. The process can be inhibited effectively by FAK antisense ODNs.


Assuntos
Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fibronectinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Animais , Células Cultivadas , DNA Antissenso/genética , DNA Antissenso/fisiologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Músculo Liso Vascular/citologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Ratos , Transfecção
18.
Yao Xue Xue Bao ; 37(5): 334-8, 2002 May.
Artigo em Zh | MEDLINE | ID: mdl-12579835

RESUMO

AIM: To study the effects of FAK-ERK1/2 signaling pathway and FAK antisense oligodeoxynucleotides (ODNs) on vascular smooth muscle cell (SMC) migration and adhesion stimulated by fibronectin (FN). METHODS: Migration and adhesion of cultured SMCs were stimulated by different concentrations of FN, FAK, ERK1/2. And their phosphorylation were detected by immunoprecipitation and Western blot. FAK antisense ODNs were transfected into SMCs by cationic lipid to investigate its modulatory effects on tyrosine phosphorylation, SMCs migration and adhesion were also measured by modifing Boyden Chamber and morphological enumeration, respectively. RESULTS: FAK were expressed when SMCs adhesion and migration were successfully simulated by FN (5, 10, 20, 40, 60 micrograms.mL-1), high contents of FAK and ERK1/2 phosphorylation were detected by 20 micrograms.mL-1 FN or more. FAK antisense ODNs were transfected efficiently by cationic lipid. FAK and ERK1/2 phosphorylation were inhibited magnificently after FAK antisense ODNs transfection. Cell migration stimulated by FN 10, 20, 40 and 60 micrograms.mL-1 were reduced by 23.26%, 21.63%, 19.31% and 17.88% respectively (P < 0.05). SMCs adhesive spreading in 5-60 micrograms.mL-1 FN groups were reduced by 17.89%-27.67% (P < 0.05). CONCLUSION: FAK-ERK1/2 mediated signal transduction play important roles in SMCs migration and adhesion stimulated by extracellular matrix. The process can be inhibited by FAK antisense ODNs effectively.


Assuntos
Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais , Animais , Aorta/citologia , Células Cultivadas , Fibronectinas/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Proteína Quinase 1 Ativada por Mitógeno/biossíntese , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Proteínas Tirosina Quinases/genética , Ratos , Ratos Sprague-Dawley , Transfecção
19.
Zhonghua Yi Xue Za Zhi ; 82(9): 622-5, 2002 May 10.
Artigo em Zh | MEDLINE | ID: mdl-12133485

RESUMO

OBJECTIVE: To study the roles of focal adhesion kinase (FAK) and mitogen activated protein kinase (p42/44MAPK) in the process of migration and proliferation of vascular smooth muscle cells (VSMC) stimulated by fibronectin (FN). METHODS: VSMCs were taken from the tunica media of SD rats and cultured. Migration and proliferation of cultured VSMCs were stimulated by different concentrations of FN; FAK, p42/44MAPK and their phosphorylation were detected by immunoprecipitation and Western blot. FAK antisense oligodeoxynucleotide (ODN) was transfected into VSMCs by cationic lipid to investigate its modulatory effects on tyrosine phosphorylation. VSMC migration and proliferation were also measured by modified Boyden Chamber and (3)H-thymidine respectively. RESULTS: FAK and p42/44MAPK were expressed when VSMC adhesion and migration were successfully simulated by FN (5, 10, 20, 40, and 60 microg/ml), high contents of FAK and p42/44MAPK phosphorylation were detected in groups with 20 microgram/ml FN or more. FAK antisense ODN was transfected efficiently by cationic lipid. Phosphorylation of FAK and p42/44MAPK was inhibited significantly after FAK antisense ODN transfection. Cell migration stimulated by FN of the concentrations of 10, 20, 40, and 60 microg/ml was reduced by 23.26%, 21.63%, 19.31% and 17.88% respectively (P < 0.05). VSMC proliferation in 5 approximately 60 microgram/ml FN groups was reduced by 27.67% approximately 46.67% (P < 0.05). CONCLUSION: FAK and p42/44MAPK play an important role in VSMC migration and proliferation stimulated by extracellular matrix. The process can be inhibited by FAK antisense ODN effectively.


Assuntos
Movimento Celular/fisiologia , Fibronectinas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Proteínas Tirosina Quinases/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Técnicas In Vitro , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Ratos , Ratos Sprague-Dawley
20.
J Alzheimers Dis ; 38(1): 171-84, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23948934

RESUMO

Aggregates of hyperphosphorylated tau (PHF-tau), such as neurofibrillary tangles, are linked to the degree of cognitive impairment in Alzheimer's disease. We have recently reported early clinical results of a novel PHF-tau targeting PET imaging agent, [F18]-T807. Since then, we have investigated a second novel PHF-tau targeting PET imaging agent, [F18]-T808, with different pharmacokinetic characteristics, which may be favorable for imaging Alzheimer's disease and other tauopathies. Here, we describe the first human brain images with [F18]-T808.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Benzimidazóis , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Fluordesoxiglucose F18 , Pirimidinas , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Benzimidazóis/farmacocinética , Encéfalo/efeitos dos fármacos , Mapeamento Encefálico , Feminino , Humanos , Masculino , Entrevista Psiquiátrica Padronizada , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Pirimidinas/farmacocinética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA