Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Circ Res ; 117(4): 376-87, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26129975

RESUMO

RATIONALE: Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function. OBJECTIVE: To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function. METHODS AND RESULTS: We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo. CONCLUSIONS: We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis.


Assuntos
Plaquetas/enzimologia , Lisofosfolipídeos/sangue , Fosfotransferases (Aceptor do Grupo Álcool)/sangue , Agregação Plaquetária , Esfingosina/análogos & derivados , Animais , Ácido Araquidônico/sangue , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/enzimologia , Modelos Animais de Doenças , Eritrócitos/enzimologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Adesividade Plaquetária , Testes de Função Plaquetária , Receptores de Lisoesfingolipídeo/sangue , Transdução de Sinais , Esfingosina/sangue , Receptores de Esfingosina-1-Fosfato , Trombose/sangue , Trombose/enzimologia , Trombose/prevenção & controle , Tromboxano A2/sangue , Lesões do Sistema Vascular/sangue , Lesões do Sistema Vascular/enzimologia
2.
Am J Physiol Renal Physiol ; 305(11): F1593-602, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23926182

RESUMO

The aims of this study were to 1) determine whether renal localization of aliskiren and its antihypertensive and renoprotective effects persist after administration of the drug is stopped and 2) define the renal localization of aliskiren by light microscopy autoradiography. Hypertensive double transgenic rats (dTGR) overexpressing genes for human renin and angiotensinogen were treated with aliskiren (3 mg·kg(-1)·day(-1) sc; osmotic minipumps) or enalapril (18 mg/l in drinking water). After a 2-wk treatment, dTGR were assigned to either continued treatment with aliskiren ("continued"), or to cessation of their respective treatment ("stopped") for a 3-wk washout. One week of treatment with aliskiren and enalapril reduced blood pressure and albuminuria vs. baseline. After cessation of either treatment, blood pressure had returned to pretreatment levels and albuminuria remained relatively low for 1 wk, but rose thereafter similarly in both groups. In contrast, renal mRNA for transforming growth factor-ß and renal collagen IV was reduced by aliskiren (continued and stopped groups), but not after cessation of enalapril. Similar patterns were found for collagen IV protein expression. Even 3 wk after stopping aliskiren treatment, renal levels of the drug exceeded its IC50, whereas enalaprilat was not detected. To localize aliskiren accumulation, Wistar rats were treated with [(3)H]-aliskiren for 7 days. Autoradiography demonstrated specific labeling in glomeruli, arterioles, and afferent arterioles as well as in the distal nephron. Labeling could still be observed even after 7 days' washout. These results suggest that the renophilic properties of aliskiren are different from enalapril and could have contributed to the renoprotective mechanism of this renin inhibitor.


Assuntos
Amidas/farmacologia , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Fumaratos/farmacologia , Rim/efeitos dos fármacos , Renina/antagonistas & inibidores , Albuminúria/metabolismo , Animais , Rim/metabolismo , Masculino , Ratos , Ratos Transgênicos , Ratos Wistar , Renina/metabolismo
3.
Hum Mol Genet ; 20(21): 4209-23, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21828077

RESUMO

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause late-onset Parkinson's disease (PD), but the underlying pathophysiological mechanisms and the normal function of this large multidomain protein remain speculative. To address the role of this protein in vivo, we generated three different LRRK2 mutant mouse lines. Mice completely lacking the LRRK2 protein (knock-out, KO) showed an early-onset (age 6 weeks) marked increase in number and size of secondary lysosomes in kidney proximal tubule cells and lamellar bodies in lung type II cells. Mice expressing a LRRK2 kinase-dead (KD) mutant from the endogenous locus displayed similar early-onset pathophysiological changes in kidney but not lung. KD mutants had dramatically reduced full-length LRRK2 protein levels in the kidney and this genetic effect was mimicked pharmacologically in wild-type mice treated with a LRRK2-selective kinase inhibitor. Knock-in (KI) mice expressing the G2019S PD-associated mutation that increases LRRK2 kinase activity showed none of the LRRK2 protein level and histopathological changes observed in KD and KO mice. The autophagy marker LC3 remained unchanged but kidney mTOR and TCS2 protein levels decreased in KD and increased in KO and KI mice. Unexpectedly, KO and KI mice suffered from diastolic hypertension opposed to normal blood pressure in KD mice. Our findings demonstrate a role for LRRK2 in kidney and lung physiology and further show that LRRK2 kinase function affects LRRK2 protein steady-state levels thereby altering putative scaffold/GTPase activity. These novel aspects of peripheral LRRK2 biology critically impact ongoing attempts to develop LRRK2 selective kinase inhibitors as therapeutics for PD.


Assuntos
Homeostase , Rim/enzimologia , Pulmão/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/ultraestrutura , Animais , Pressão Sanguínea/efeitos dos fármacos , Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Estabilidade Enzimática/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Rim/ultraestrutura , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Túbulos Renais Proximais/fisiopatologia , Túbulos Renais Proximais/ultraestrutura , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Mutantes , Atividade Motora , Transdução de Sinais/efeitos dos fármacos
4.
J Clin Invest ; 110(12): 1869-77, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12488437

RESUMO

Preformed and elicited Ab's against the Galalpha1,3Gal terminating carbohydrate chains (alphaGal Ab's) are the primary cause of hyperacute and acute vascular xenograft rejection in pig-to-primate transplantation. alphaGal Ab's are produced by long-lived Ab-producing cells that are not susceptible to pharmacological immunosuppression. We reasoned that antigen-specific elimination of alphaGal Ab's might be achieved in vivo by systemic administration of nonimmunogenic polyvalent alphaGal structures with high avidity for alphaGal Ab's. We devised GAS914, a soluble trisaccharide-polylysine conjugate of approximately 500 kDa that effectively competes for alphaGal binding by alphaGal IgM (IC(50), 43 nM) and IgG (IC(50), 28 nM) in vitro. Injections of GAS914 in cynomolgus monkeys, at the dose of 1 mg/kg, resulted in the immediate decrease of more than 90% of circulating alphaGal Ab's and serum anti-pig cytotoxicity. In baboons, repeated injections of GAS914 effectively reduced both circulating alphaGal Ab's and cytotoxicity over several months. Studies with [(14)C]GAS914 in rhesus monkeys and Gal(-/-) mice indicate that GAS914 binds to circulating alphaGal Ab's and that the complex is quickly metabolized by the liver and excreted by the kidney. Remarkably, posttreatment alphaGal Ab titers never exceeded pretreatment levels and no sensitization to either alphaGal or the polylysine backbone has been observed. Furthermore there was no apparent acute or chronic toxicity associated with GAS914 treatment in primates. We conclude that GAS914 may be used therapeutically for the specific removal of alphaGal Ab's.


Assuntos
Anticorpos Heterófilos/sangue , Dissacarídeos/imunologia , Epitopos/imunologia , Técnicas de Imunoadsorção , Polímeros/administração & dosagem , Trissacarídeos/administração & dosagem , Animais , Anticorpos Heterófilos/imunologia , Autorradiografia/métodos , Radioisótopos de Carbono/química , Radioisótopos de Carbono/metabolismo , Dissacarídeos/genética , Dissacarídeos/metabolismo , Epitopos/genética , Epitopos/metabolismo , Feminino , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Humanos , Macaca fascicularis , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Estrutura Molecular , Papio , Polímeros/química , Ratos , Suínos , Distribuição Tecidual , Imunologia de Transplantes , Transplante Heterólogo , Trissacarídeos/genética , Trissacarídeos/imunologia , Trissacarídeos/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-16898071

RESUMO

ASM 981 has been developed for topical treatment of inflammatory skin diseases. It specifically inhibits the production and release of pro-inflammatory cytokines. We measured the skin penetration of ASM 981 in canine skin and compared penetration in living and frozen skin. To make penetration of ASM 981 visible in dog skin, tritium labelled ASM 981 was applied to a living dog and to defrosted skin of the same dog. Using qualitative autoradiography the radioactive molecules were detected in the lumen of the hair follicles until the infundibulum, around the superficial parts of the hair follicles and into a depth of the dermis of 200 to 500 microm. Activity could not be found in deeper parts of the hair follicles, the dermis or in the sebaceous glands. Penetration of ASM 981 is low in canine skin and is only equally spread in the upper third of the dermis 24 hours after application. Penetration in frozen skin takes even longer than in living canine skin but shows the same distribution.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Criopreservação , Absorção Cutânea , Pele/metabolismo , Tacrolimo/análogos & derivados , Administração Cutânea , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Pele/anatomia & histologia , Tacrolimo/administração & dosagem , Tacrolimo/metabolismo , Fatores de Tempo
6.
Eur J Cancer ; 41(10): 1460-6, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15913989

RESUMO

The experimentally induced neurotoxic effects of paclitaxel and docetaxel have never been compared, since no animal models of docetaxel peripheral neurotoxicity have yet been reported. In this experiment, we examined the effect of the chronic administration of these two taxanes in the Wistar rat using neurophysiological, neuropathological and morphometrical methods. Our results showed that both paclitaxel and docetaxel induced a significant, equally severe and dose-dependent reduction in nerve conduction velocity. On the contrary, the morphometric examination demonstrated that the effect on the nerve fibres was more severe after paclitaxel administration when the same schedule was used. However, the overall severity of the pathological changes was milder than expected on the basis of the neurophysiological results. Our results support the hypothesis that taxanes (and particularly docetaxel) may exert their neurotoxic effect not only on the microtubular system of the peripheral nerves, but also on other less obvious targets.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Paclitaxel/efeitos adversos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Taxoides/efeitos adversos , Análise de Variância , Animais , Docetaxel , Feminino , Microtúbulos/patologia , Fibras Nervosas Mielinizadas/patologia , Fator de Crescimento Neural/sangue , Doenças do Sistema Nervoso Periférico/patologia , Ratos , Ratos Wistar
7.
Endocr Pathol ; 14(1): 81-91, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12746566

RESUMO

Spontaneously occurring and chemically induced pheochromocytomas are rare in mice. That the mouse pheochromocytoma is a more appropriate animal model than that of the rat for study of human medullary adrenal tumors has been suggested. The expression of phenylethanolamine-N-methyltransferase (PNMT), the enzyme responsible for production of epinephrine from norepinephrine, is common to both mouse and human pheochromocytomas. This investigation assessed the expression of the immunohistochemical markers PNMT, tyrosine hydroxylase (TH), and chromogranin A (CGA) in spontaneously occurring and chemically induced pheochromocytomas in the B6C3F1 mouse. Spontaneous tumors were derived from control animals from 10 different studies and the pheochromocytomas from treated groups from 4 different studies. All tumors were positive for maximal TH expression. A highly significant difference in PNMT expression (p < 0.01) occurred between spontaneously occurring pheochromocytomas classified as benign or "malignant" by the criteria of toxicologic pathology. Chemically induced tumors showed intermediate PNMT staining. A marked reduction in CGA expression occurred in pheochromocytomas induced by technical grade pentachlorophenol, compared to the other three chemicals and the spontaneously occurring tumors. These findings suggest that immunohistochemistry is a reliable tool in investigating the functional capabilities of pheochromocytomas in mice. PNMT expression is a tightly regulated component of the chromaffin cell phenotype and appears to be readily lost in mouse pheochromocytomas, particularly those with aggressive characteristics.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Cromograninas/biossíntese , Feniletanolamina N-Metiltransferase/biossíntese , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Tirosina 3-Mono-Oxigenase/biossíntese , Neoplasias das Glândulas Suprarrenais/induzido quimicamente , Animais , Cromogranina A , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Neoplasias , Feocromocitoma/induzido quimicamente , Ratos
8.
Mol Cell Biol ; 32(14): 2871-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22586266

RESUMO

Brown adipose tissue (BAT) is a key tissue for energy expenditure via fat and glucose oxidation for thermogenesis. In this study, we demonstrate that the myostatin/activin receptor IIB (ActRIIB) pathway, which serves as an important negative regulator of muscle growth, is also a negative regulator of brown adipocyte differentiation. In parallel to the anticipated hypertrophy of skeletal muscle, the pharmacological inhibition of ActRIIB in mice, using a neutralizing antibody, increases the amount of BAT without directly affecting white adipose tissue. Mechanistically, inhibition of ActRIIB inhibits Smad3 signaling and activates the expression of myoglobin and PGC-1 coregulators in brown adipocytes. Consequently, ActRIIB blockade in brown adipose tissue enhances mitochondrial function and uncoupled respiration, translating into beneficial functional consequences, including enhanced cold tolerance and increased energy expenditure. Importantly, ActRIIB inhibition enhanced energy expenditure only at ambient temperature or in the cold and not at thermoneutrality, where nonshivering thermogenesis is minimal, strongly suggesting that brown fat activation plays a prominent role in the metabolic actions of ActRIIB inhibition.


Assuntos
Receptores de Activinas Tipo II/antagonistas & inibidores , Adipogenia/fisiologia , Tecido Adiposo Marrom/metabolismo , Termogênese/fisiologia , Receptores de Activinas Tipo II/imunologia , Receptores de Activinas Tipo II/metabolismo , Adipócitos Marrons/citologia , Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/ultraestrutura , Animais , Anticorpos Neutralizantes , Diferenciação Celular , Metabolismo Energético , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Fatores de Transcrição/metabolismo
9.
Leuk Res ; 35(5): 631-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21129774

RESUMO

In vitro, concentrations ≥ 10 µM of nilotinib were needed to induce markers of cytotoxicity, apoptosis, and endoplasmic reticulum stress in both neonatal rat ventricular myocytes, a putative target tissue, and non-target heart fibroblasts, indicating a lack of cardiomyocyte-specific nilotinib toxicity in vitro. In rats, oral nilotinib treatment at 80 mg/kg for 4 weeks induced increased heart weight; however, this was not associated with relevant histopathological changes or effects on heart function. Thus, nilotinib at and above clinically relevant concentrations (4.27 µM) did not induce overt cardiovascular pathologies or heart failure in vitro or in vivo under study conditions.


Assuntos
Cardiotoxinas , Coração/efeitos dos fármacos , Pirimidinas/efeitos adversos , Animais , Animais Recém-Nascidos , Cardiotoxinas/efeitos adversos , Cardiotoxinas/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Coração/fisiologia , Ventrículos do Coração/anatomia & histologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/ultraestrutura , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Pirimidinas/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Função Ventricular/efeitos dos fármacos
10.
Leuk Res ; 34(9): 1180-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20122731

RESUMO

Cytotoxic concentrations of imatinib mesylate (10-50 microM) were required to trigger markers of apoptosis and endoplasmic reticulum stress response in neonatal rat ventricular myocytes and fibroblasts, with no significant differences observed between c-Abl silenced and nonsilenced cells. In mice, oral or intraperitoneal imatinib treatment did not induce cardiovascular pathology or heart failure. In rats, high doses of oral imatinib did result in some cardiac hypertrophy. Multi-organ toxicities may have increased the cardiac workload and contributed to the cardiac hypertrophy observed in rats only. These data suggest that imatinib is not cardiotoxic at clinically relevant concentrations (5 microM).


Assuntos
Antineoplásicos/efeitos adversos , Coração/efeitos dos fármacos , Piperazinas/efeitos adversos , Pirimidinas/efeitos adversos , Animais , Sequência de Bases , Benzamidas , Primers do DNA , Coração/fisiologia , Mesilato de Imatinib , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar
11.
Hypertension ; 52(1): 130-6, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18490518

RESUMO

The aim of this study was to explore the effects of the renin inhibitor aliskiren in streptozotocin-diabetic TG(mRen-2)27 rats. Furthermore, we investigated in vitro the effect of aliskiren on the interactions between renin and the (pro)renin receptor and between aliskiren and prorenin. Aliskiren distributed extensively to the kidneys of normotensive (non)diabetic rats, localizing in the glomeruli and vessel walls after 2 hours exposure. In diabetic TG(mRen-2)27 rats, aliskiren (10 or 30 mg/kg per day, 10 weeks) lowered blood pressure, prevented albuminuria, and suppressed renal transforming growth factor-beta and collagen I expression versus vehicle. Aliskiren reduced (pro)renin receptor expression in glomeruli, tubules, and cortical vessels compared to vehicle (in situ hybridization). In human mesangial cells, aliskiren (0.1 micromol/L to 10 micromol/L) did not inhibit binding of (125)I-renin to the (pro)renin receptor, nor did it alter the activation of extracellular signal-regulated kinase 1/2 by renin (20 nmol/L) preincubated with aliskiren (100 nmol/L) or affect gene expression of the (pro)renin receptor. Evidence was obtained that aliskiren binds to the active site of prorenin. The above results demonstrate the antihypertensive and renoprotective effects of aliskiren in experimental diabetic nephropathy. The evidence that aliskiren can reduce in vivo gene expression for the (pro)renin receptor and that it may block prorenin-induced angiotensin generation supports the need for additional work to reveal the mechanism of the observed renoprotection by this renin inhibitor.


Assuntos
Albuminúria/fisiopatologia , Amidas/farmacologia , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/fisiopatologia , Fumaratos/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Renina/antagonistas & inibidores , Albuminúria/etiologia , Albuminúria/metabolismo , Amidas/farmacocinética , Animais , Anti-Hipertensivos/metabolismo , Anti-Hipertensivos/farmacocinética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/metabolismo , Fumaratos/farmacocinética , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Renina/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Receptor de Pró-Renina
12.
J Pharmacol Exp Ther ; 323(2): 469-75, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17682127

RESUMO

FTY720 [2-amino-2-[2-(4-octylphenyl) ethyl]propane-1,3-diol hydrochloride] is an oral sphingosine-1-phosphate receptor modulator under development for the treatment of multiple sclerosis (MS). The drug is phosphorylated in vivo by sphingosine kinase 2 to its bioactive form, FTY720-P. Although treatment with FTY720 is accompanied by a reduction of the peripheral lymphocyte count, its efficacy in MS and experimental autoimmune encephalomyelitis (EAE) may be due to additional, direct effects in the central nervous system (CNS). We now show that FTY720 localizes to the CNS white matter, preferentially along myelin sheaths. Brain trough levels of FTY720 and FTY720-P in rat EAE are of the same magnitude and dose dependently increase; they are in the range of 40 to 540 ng/g in the brain tissue at efficacious doses and exceed blood concentrations severalfold. In a rat model of chronic EAE, prolonged treatment with 0.03 mg/kg was efficacious, but limiting the dosing period failed to prevent EAE despite a significant decrease in blood lymphocytes. FTY720 effectiveness is likely due to a culmination of mechanisms involving reduction of autoreactive T cells, neuroprotective influence of FTY720-P in the CNS, and inhibition of inflammatory mediators in the brain.


Assuntos
Encéfalo/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Imunossupressores/farmacocinética , Esclerose Múltipla/tratamento farmacológico , Propilenoglicóis/farmacocinética , Esfingosina/análogos & derivados , Administração Oral , Animais , Autorradiografia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Cloridrato de Fingolimode , Contagem de Linfócitos , Fosforilação , Propilenoglicóis/administração & dosagem , Propilenoglicóis/uso terapêutico , Ratos , Ratos Endogâmicos Lew , Esfingosina/administração & dosagem , Esfingosina/farmacocinética , Esfingosina/uso terapêutico
13.
Radiology ; 234(3): 765-75, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15665219

RESUMO

PURPOSE: To describe dose-dependent signal intensity (SI) characteristics of experimentally induced soft-tissue abscesses on 1.5-T T1- and T2*-weighted magnetic resonance (MR) images obtained 24 hours after administration of ultrasmall superparamagnetic iron oxide (USPIO) and to describe the relationship between SI and amount of USPIO uptake and macrophage iron content. MATERIALS AND METHODS: Local institutional review committee on animal care approved the experiments, which were performed according to the guidelines of the National Institutes of Health and the committee on animal research at our institution. Unilateral calf muscle abscesses were induced in 21 rats with an injection of a Staphylococcus aureus suspension. The rats were divided into three groups of seven animals each: low USPIO dose (50 micromol of iron per kilogram of body weight), high USPIO dose (150 micromol Fe/kg), and control (saline solution). All rats were imaged before and 24 hours after USPIO administration at 1.5 T (transverse T1-weighted spin-echo, T2*-weighted fast gradient-echo, and short inversion time inversion-recovery sequences). Images were analyzed quantitatively and qualitatively with regard to SI and signal pattern. Temporal variation of calculated contrast-to-noise ratios was analyzed with the Wilcoxon signed rank test. MR findings were correlated with histopathologic findings, including those of electron microscopy. RESULTS: Twenty-four hours after USPIO administration in the high-dose group, susceptibility effects were present in abscess periphery on postcontrast T2*-weighted images (P=.04), and SI enhancement was noted on postcontrast T1-weighted images within both abscess wall and abscess center (P=.04 for both). In the low-dose group, SI enhancement was noted in entire abscess on T1-weighted postcontrast images (P=.03). Neither significant SI loss (P=.09) nor susceptibility effects were detected in periphery or center of any abscess on postcontrast T2*-weighted images. There was no obvious difference in total amount of macrophages among the groups, but there was a clear difference with regard to individual iron content of iron-positive macrophages between the USPIO dose groups. CONCLUSION: At 1.5 T, SI characteristics of abscesses on T1- and T2*-weighted images obtained 24 hours after USPIO injection strongly depend on administered dose of the contrast agent. At low doses, T1 effects were stronger than T2* effects.


Assuntos
Abscesso/patologia , Meios de Contraste/metabolismo , Ferro/metabolismo , Macrófagos/metabolismo , Imageamento por Ressonância Magnética/métodos , Óxidos/metabolismo , Infecções dos Tecidos Moles/patologia , Infecções Estafilocócicas/patologia , Abscesso/imunologia , Animais , Meios de Contraste/administração & dosagem , Dextranos , Feminino , Óxido Ferroso-Férrico , Ferro/administração & dosagem , Ativação de Macrófagos , Nanopartículas de Magnetita , Óxidos/administração & dosagem , Projetos Piloto , Ratos , Ratos Sprague-Dawley , Infecções dos Tecidos Moles/imunologia , Infecções Estafilocócicas/imunologia , Estatísticas não Paramétricas
14.
J Cardiovasc Pharmacol ; 40(5): 789-800, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12409988

RESUMO

Combination of nonhypotensive doses of valsartan and enalapril markedly improved survival (+87%) compared with untreated animals (37%) in spontaneously hypertensive rats (SHRs) with endothelial dysfunction. However, the combination had no effect on kidney function, and proteinuria persisted over the 12 weeks of the study. It was hypothesized that the greater survival was due to improvement in endothelial function or coronary vasculature despite blockade of nitric oxide synthase and high blood pressure. Therefore, endothelial function was evaluated in isolated aortic ring and maximal coronary blood flow was studied in isolated perfused SHR hearts (20-24 weeks) treated with -nitro-l-arginine methyl ester (L-NAME) (50 mg/l) for 4 weeks. The animals received vehicle, valsartan 5 mg/kg/d, enalapril 1 mg/kg/d, valsartan 50 mg/kg/d, or the combination valsartan 5 mg/kg/d with enalapril 1 mg/kg/d in drinking water. Normotensive Wistar-Kyoto (WKY) rats were used as control. Blood pressure was measured by telemetry. Histopathology was performed on heart, kidney (hematoxylin-eosin), and aorta (Masson trichrome). L-NAME elevated blood pressure by 50 mm Hg after vehicle (199 +/- 5 mm Hg). Valsartan 50 mg/kg/d completely abolished this increase (150 +/- 4 mm Hg) whereas the valsartan-enalapril combination synergistically decreased blood pressure (-37 mm Hg at 162 +/- 7 mm Hg) compared with monotherapy (valsartan 5 mg/kg/d -10 mm Hg; enalapril 1 mg/kg/d -15 mm Hg). All treatments improved the histopathology, most markedly in those receiving the valsartan-enalapril combination. The severity mean grades for lesions were 2.1, 1.9, 1.7, 1.1, and 0.9 in vehicle-treated SHRs, enalapril 1 mg/kg/d, valsartan 5 mg/kg/d, valsartan 5 or 50 mg/kg/d, and the valsartan-enalapril combination, respectively, compared with 0.02 in WKY rats. Acetylcholine-induced relaxation was significantly greater in treated SHRs than after vehicle (-40% at 0.1 mmol acetylcholine) but the combination induced the maximal relaxation (-85%). The ratio of maximal tension induced by serotonin in rings with and without endothelium was 1.4 and 1.3 in vehicle and valsartan 5 mg/kg/d-treated rats whereas it did not differ from 1 in WKY rats and all other treated groups. The cardiac hypertrophy (+27%) was prevented by valsartan 50 mg/kg/d and the valsartan-enalapril combination. Coronary reserve was significantly increased by valsartan 50 mg/kg/d (+85% at 7.8 +/- 0.7 ml/min/g) and the valsartan-enalapril combination (+42% at 6.0 +/- 0.4 ml/min/g) compared with 4.2 +/- 0.5 (vehicle). This was not different of 8.8 +/- 0.5 (WKYs). Despite the maintenance of a high blood pressure, low-dose valsartan-enalapril significantly improved endothelial function and histopathology and increased coronary reserve in SHRs chronically receiving L-NAME.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Enalapril/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Tetrazóis/uso terapêutico , Valina/análogos & derivados , Valina/uso terapêutico , Animais , Circulação Coronária , Quimioterapia Combinada , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Valsartana
15.
An. R. Acad. Farm ; 66(4): 541-572, oct. 2000. ilus, tab, graf
Artigo em En | IBECS (Espanha) | ID: ibc-17215

RESUMO

La cefalosporina A (CsA) se utiliza frecuentemente en hepatocitos de rata y fracciones mitocondriales hepáticas aisladas como inhibidor específico de la liberación de Ca2+ y como bloqueador selectivo de la permeabilidad y del potencial de membrana mitocondriales, procesos implicados en la inhibición de la apoptosis. Sin embargo, hasta ahora no se ha descrito ni la inhibición ni la inducción de apoptosis por CsA en cultivos primarios de hepatocitos de rata tras su incubación por un periodo de 4 a 20 horas. El propósito de este estudio ha sido examinar con criterios morfológicos y bioquímicos los efectos de la CsA sobre la apoptosis y esclarecer las características de los mecanismos subyacentes. Los hepatocitos de rata se cultivaron durante 4-20 horas con CsA a concentraciones de 0, 10, 25 y 50 µM. Se investigaron los fenómenos de condensación y fragmentación de la cromatina, fragmentación de ADN (TUNEL), distribución de fosfatidilcolina en la membrana (Anexina V), así como la actividad enzimática de caspasas 1, -3 y -6, el potencial de membrana mitocondrial (Rhodamina 123) y la liberación de citocromo C en el citosol. Tras cuatro horas de incubación con CsA, la condensación y fragmentación de la cromatina y el número de células TUNEL y Anexina V positivas aumentaron en función de la dosis sin que se observara pérdida enzimática, lo que indica la integridad de la membrana celular externa. Después de 20 horas de incubación con CsA, experimentaron unmayor incremento acompañado del aumento de las actividad de cistein-proteasa, caspasa-3 (CPP32) y de un ligero incremento de caspasa-6 (Mch 2), pero no de caspasa-1 (ICE). El inhibidor de caspasa-6, Ac-VEID-CHO, únicamente inhibió la apoptosis inducida por CsA. El descenso de potencial de membrana mitocondrial y la liberación de citocromo C fueron paralelos a los cambios de ultraestructuras mitocondriales y pudieran considerarse reacciones tempranas que desencadenan la cascada de fenómenos apoptóticos. La microscopía de transmisión electrónica (TEM) confirmó el incremento del número de células necróticas al cabo de 20 horas, pero no tras 4 horas de incubación, en comparación con los controles. (AU)


Assuntos
Animais , Masculino , Ratos , Apoptose , Ciclosporina/farmacologia , Imunossupressores/farmacologia , Fígado/citologia , Fígado , Microscopia Eletrônica , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa