Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Biol Chem ; 298(12): 102625, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36306823

RESUMO

Mucopolysaccharidosis type IIIA (MPS IIIA) is a lysosomal storage disorder caused by N-sulfoglucosamine sulfohydrolase (SGSH) deficiency. SGSH removes the sulfate from N-sulfoglucosamine residues on the nonreducing end of heparan sulfate (HS-NRE) within lysosomes. Enzyme deficiency results in accumulation of partially degraded HS within lysosomes throughout the body, leading to a progressive severe neurological disease. Enzyme replacement therapy has been proposed, but further evaluation of the treatment strategy is needed. Here, we used Chinese hamster ovary cells to produce a highly soluble and fully active recombinant human sulfamidase (rhSGSH). We discovered that rhSGSH utilizes both the CI-MPR and LRP1 receptors for uptake into patient fibroblasts. A single intracerebroventricular (ICV) injection of rhSGSH in MPS IIIA mice resulted in a tissue half-life of 9 days and widespread distribution throughout the brain. Following a single ICV dose, both total HS and the MPS IIIA disease-specific HS-NRE were dramatically reduced, reaching a nadir 2 weeks post dose. The durability of effect for reduction of both substrate and protein markers of lysosomal dysfunction and a neuroimmune response lasted through the 56 days tested. Furthermore, seven weekly 148 µg doses ICV reduced those markers to near normal and produced a 99.5% reduction in HS-NRE levels. A pilot study utilizing every other week dosing in two animals supports further evaluation of less frequent dosing. Finally, our dose-response study also suggests lower doses may be efficacious. Our findings show that rhSGSH can normalize lysosomal HS storage and markers of a neuroimmune response when delivered ICV.


Assuntos
Encefalopatias , Mucopolissacaridose III , Cricetinae , Animais , Humanos , Camundongos , Mucopolissacaridose III/tratamento farmacológico , Mucopolissacaridose III/metabolismo , Células CHO , Projetos Piloto , Cricetulus , Hidrolases/metabolismo , Encéfalo/metabolismo , Heparitina Sulfato/metabolismo , Encefalopatias/metabolismo , Lisossomos/metabolismo , Modelos Animais de Doenças
2.
J Biol Chem ; 295(39): 13556-13569, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32727849

RESUMO

Mutations in the galactosidase ß 1 (GLB1) gene cause lysosomal ß-galactosidase (ß-Gal) deficiency and clinical onset of the neurodegenerative lysosomal storage disease, GM1 gangliosidosis. ß-Gal and neuraminidase 1 (NEU1) form a multienzyme complex in lysosomes along with the molecular chaperone, protective protein cathepsin A (PPCA). NEU1 is deficient in the neurodegenerative lysosomal storage disease sialidosis, and its targeting to and stability in lysosomes strictly depend on PPCA. In contrast, ß-Gal only partially depends on PPCA, prompting us to investigate the role that ß-Gal plays in the multienzyme complex. Here, we demonstrate that ß-Gal negatively regulates NEU1 levels in lysosomes by competitively displacing this labile sialidase from PPCA. Chronic cellular uptake of purified recombinant human ß-Gal (rhß-Gal) or chronic lentiviral-mediated GLB1 overexpression in GM1 gangliosidosis patient fibroblasts coincides with profound secondary NEU1 deficiency. A regimen of intermittent enzyme replacement therapy dosing with rhß-Gal, followed by enzyme withdrawal, is sufficient to augment ß-Gal activity levels in GM1 gangliosidosis patient fibroblasts without promoting NEU1 deficiency. In the absence of ß-Gal, NEU1 levels are elevated in the GM1 gangliosidosis mouse brain, which are restored to normal levels following weekly intracerebroventricular dosing with rhß-Gal. Collectively, our results highlight the need to carefully titrate the dose and dosing frequency of ß-Gal augmentation therapy for GM1 gangliosidosis. They further suggest that intermittent intracerebroventricular enzyme replacement therapy dosing with rhß-Gal is a tunable approach that can safely augment ß-Gal levels while maintaining NEU1 at physiological levels in the GM1 gangliosidosis brain.


Assuntos
Terapia de Reposição de Enzimas , Fibroblastos/enzimologia , Lisossomos/enzimologia , Mucolipidoses , beta-Galactosidase/uso terapêutico , Animais , Células CHO , Cricetulus , Humanos , Lisossomos/genética , Camundongos , Camundongos Mutantes , Mucolipidoses/tratamento farmacológico , Mucolipidoses/enzimologia , Mucolipidoses/genética , Neuraminidase/genética , Neuraminidase/metabolismo
3.
J Biol Chem ; 295(39): 13532-13555, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31481471

RESUMO

Autosomal recessive mutations in the galactosidase ß1 (GLB1) gene cause lysosomal ß-gal deficiency, resulting in accumulation of galactose-containing substrates and onset of the progressive and fatal neurodegenerative lysosomal storage disease, GM1 gangliosidosis. Here, an enzyme replacement therapy (ERT) approach in fibroblasts from GM1 gangliosidosis patients with recombinant human ß-gal (rhß-gal) produced in Chinese hamster ovary cells enabled direct and precise rhß-gal delivery to acidified lysosomes. A single, low dose (3 nm) of rhß-gal was sufficient for normalizing ß-gal activity and mediating substrate clearance for several weeks. We found that rhß-gal uptake by the fibroblasts is dose-dependent and saturable and can be competitively inhibited by mannose 6-phosphate, suggesting cation-independent, mannose 6-phosphate receptor-mediated endocytosis from the cell surface. A single intracerebroventricularly (ICV) administered dose of rhß-gal (100 µg) resulted in broad bilateral biodistribution of rhß-gal to critical regions of pathology in a mouse model of GM1 gangliosidosis. Weekly ICV dosing of rhß-gal for 8 weeks substantially reduced brain levels of ganglioside and oligosaccharide substrates and reversed well-established secondary neuropathology. Of note, unlike with the ERT approach, chronic lentivirus-mediated GLB1 overexpression in the GM1 gangliosidosis patient fibroblasts caused accumulation of a prelysosomal pool of ß-gal, resulting in activation of the unfolded protein response and endoplasmic reticulum stress. This outcome was unsurprising in light of our in vitro biophysical findings for rhß-gal, which include pH-dependent and concentration-dependent stability and dynamic self-association. Collectively, our results highlight that ICV-ERT is an effective therapeutic intervention for managing GM1 gangliosidosis potentially more safely than with gene therapy approaches.


Assuntos
Terapia de Reposição de Enzimas , Gangliosidose GM1/terapia , beta-Galactosidase/metabolismo , Animais , Gangliosidose GM1/metabolismo , Gangliosidose GM1/patologia , Camundongos
4.
Toxicol Pathol ; 49(4): 950-962, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33691530

RESUMO

Scoring demyelination and regeneration in hematoxylin and eosin-stained nerves poses a challenge even for the trained pathologist. This article demonstrates how combinatorial multiplex immunohistochemistry (IHC) and quantitative digital pathology bring new insights into the peripheral neuropathogenesis of the Twitcher mouse, a model of Krabbe disease. The goal of this investigational study was to integrate modern pathology tools to traditional anatomic pathology microscopy workflows, in order to generate quantitative data in a large number of samples, and aid the understanding of complex disease pathomechanisms. We developed a novel IHC toolkit using a combination of CD68, periaxin-1, phosphorylated neurofilaments and SOX-10 to interrogate inflammation, myelination, axonal size, and Schwann cell counts in sciatic nerves from 17-, 21-, 25-, and 35-day-old wild-type and Twitcher mice using self-customized digital image algorithms. Our quantitative analyses highlight that nerve macrophage infiltration and interstitial expansion are the earliest detectable changes in Twitcher nerves. By 17 days of age, while the diameter of axons is small, the number of myelinated axons is still normal. However, from 21 days onward Twitcher nerves contain 75% of wild-type myelinated nerve fiber numbers despite containing 3 times more Schwann cells. In 35-day-old Twitcher mice when demyelination is detectable, nerve myelination drops to 50%.


Assuntos
Leucodistrofia de Células Globoides , Nervo Isquiático , Animais , Axônios , Modelos Animais de Doenças , Camundongos , Camundongos Mutantes Neurológicos , Regeneração Nervosa
5.
Proc Natl Acad Sci U S A ; 111(41): 14870-5, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25267636

RESUMO

Mucopolysaccharidosis type IIIB (MPS IIIB, Sanfilippo syndrome type B) is a lysosomal storage disease characterized by profound intellectual disability, dementia, and a lifespan of about two decades. The cause is mutation in the gene encoding α-N-acetylglucosaminidase (NAGLU), deficiency of NAGLU, and accumulation of heparan sulfate. Impediments to enzyme replacement therapy are the absence of mannose 6-phosphate on recombinant human NAGLU and the blood-brain barrier. To overcome the first impediment, a fusion protein of recombinant NAGLU and a fragment of insulin-like growth factor II (IGFII) was prepared for endocytosis by the mannose 6-phosphate/IGFII receptor. To bypass the blood-brain barrier, the fusion protein ("enzyme") in artificial cerebrospinal fluid ("vehicle") was administered intracerebroventricularly to the brain of adult MPS IIIB mice, four times over 2 wk. The brains were analyzed 1-28 d later and compared with brains of MPS IIIB mice that received vehicle alone or control (heterozygous) mice that received vehicle. There was marked uptake of the administered enzyme in many parts of the brain, where it persisted with a half-life of approximately 10 d. Heparan sulfate, and especially disease-specific heparan sulfate, was reduced to control level. A number of secondary accumulations in neurons [ß-hexosaminidase, LAMP1(lysosome-associated membrane protein 1), SCMAS (subunit c of mitochondrial ATP synthase), glypican 5, ß-amyloid, P-tau] were reduced almost to control level. CD68, a microglial protein, was reduced halfway. A large amount of enzyme also appeared in liver cells, where it reduced heparan sulfate and ß-hexosaminidase accumulation to control levels. These results suggest the feasibility of enzyme replacement therapy for MPS IIIB.


Assuntos
Acetilglucosaminidase/uso terapêutico , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos , Fator de Crescimento Insulin-Like II/uso terapêutico , Mucopolissacaridose III/tratamento farmacológico , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Biomarcadores/metabolismo , Encéfalo/patologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Endocitose , Fibroblastos/metabolismo , Fibroblastos/patologia , Heparitina Sulfato/metabolismo , Humanos , Injeções Intraventriculares , Fígado/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , Mucopolissacaridose III/patologia , Neurônios/metabolismo , Neurônios/patologia , Ligação Proteica , beta-N-Acetil-Hexosaminidases/metabolismo
7.
Bioorg Med Chem Lett ; 21(3): 983-8, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21211973

RESUMO

1-(1-Acetyl-piperidin-4-yl)-3-adamantan-1-yl-urea 14a (AR9281), a potent and selective soluble epoxide hydrolase inhibitor, was recently tested in a phase 2a clinical setting for its effectiveness in reducing blood pressure and improving insulin resistance in pre-diabetic patients. In a mouse model of diet induced obesity, AR9281 attenuated the enhanced glucose excursion following an intraperitoneal glucose tolerance test. AR9281 also attenuated the increase in blood pressure in angiotensin-II-induced hypertension in rats. These effects were dose-dependent and well correlated with inhibition of the sEH activity in whole blood, consistent with a role of sEH in the observed pharmacology in rodents.


Assuntos
Adamantano/análogos & derivados , Anti-Hipertensivos/química , Inibidores Enzimáticos/química , Epóxido Hidrolases/antagonistas & inibidores , Hipertensão/tratamento farmacológico , Resistência à Insulina , Ureia/análogos & derivados , Adamantano/química , Adamantano/farmacocinética , Adamantano/uso terapêutico , Administração Oral , Angiotensina II/farmacologia , Animais , Anti-Hipertensivos/farmacocinética , Anti-Hipertensivos/uso terapêutico , Glicemia/análise , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/uso terapêutico , Epóxido Hidrolases/metabolismo , Hipertensão/induzido quimicamente , Camundongos , Obesidade/tratamento farmacológico , Ratos , Ureia/química , Ureia/farmacocinética , Ureia/uso terapêutico
8.
Arterioscler Thromb Vasc Biol ; 29(9): 1265-70, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19667112

RESUMO

OBJECTIVE: Epoxyeicosatrienoic acids (EETs) have been shown to have antiinflammatory effects and therefore may play a role in preventing vascular inflammatory and atherosclerotic diseases. Soluble epoxide hydrolase (s-EH) converts EETs into less bioactive dihydroxyeicosatrienoic acids. Thus, inhibition of s-EH can prevent degradation of EETs and prolong their effects. The present study aimed to test the hypothesis that inhibition of s-EH has vascular protective effects. METHODS AND RESULTS: Six-month-old apolipoprotein E-deficient mice were chronically infused with angiotensin II (1.44 mg/kg/d) for 4 weeks to induce abdominal aortic aneurysm (AAA), accelerate atherosclerosis development and carotid artery ligation-induced vascular remodeling. The mice were treated with a novel s-EH inhibitor, AR9276 (1.5 g/L in drinking water) or vehicle for 4 weeks. The results demonstrated that AR9276 significantly reduced the rate of AAA formation and atherosclerotic lesion area, but had no effect on ligation-induced carotid artery remodeling. These effects were associated with a reduction of serum lipid, IL-6, murine IL-8-KC, and IL-1alpha, and downregulation of gene expressions of ICAM-1, VCAM-1, and IL-6 in the arterial wall. CONCLUSIONS: The present data demonstrate that treatment with an s-EH inhibitor attenuates AAA formation and atherosclerosis development. The attendant downregulation of inflammatory mediators and lipid lowering effects may both contribute to the observed vascular protective effects.


Assuntos
Aneurisma da Aorta Abdominal/prevenção & controle , Aterosclerose/prevenção & controle , Dislipidemias/prevenção & controle , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Administração Oral , Angiotensina II , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/enzimologia , Aneurisma da Aorta Abdominal/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/induzido quimicamente , Aterosclerose/enzimologia , Aterosclerose/patologia , Disponibilidade Biológica , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Artérias Carótidas/cirurgia , Colesterol/sangue , Modelos Animais de Doenças , Regulação para Baixo , Dislipidemias/induzido quimicamente , Dislipidemias/enzimologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Epóxido Hidrolases/metabolismo , Mediadores da Inflamação/sangue , Molécula 1 de Adesão Intercelular/genética , Interleucina-1alfa/sangue , Interleucina-6/sangue , Interleucina-6/genética , Interleucina-8/sangue , Ligadura , Masculino , Camundongos , Camundongos Knockout , Molécula 1 de Adesão de Célula Vascular/genética
9.
Drug Deliv Transl Res ; 10(2): 425-439, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31942701

RESUMO

BMN 250 is being developed as enzyme replacement therapy for Sanfilippo type B, a primarily neurological rare disease, in which patients have deficient lysosomal alpha-N-acetylglucosaminidase (NAGLU) enzyme activity. BMN 250 is taken up in target cells by the cation-independent mannose 6-phosphate receptor (CI-MPR, insulin-like growth factor 2 receptor), which then facilitates transit to the lysosome. BMN 250 is dosed directly into the central nervous system via the intracerebroventricular (ICV) route, and the objective of this work was to compare systemic intravenous (IV) and ICV delivery of BMN 250 to confirm the value of ICV dosing. We first assess the ability of enzyme to cross a potentially compromised blood-brain barrier in the Naglu-/- mouse model and then assess the potential for CI-MPR to be employed for receptor-mediated transport across the blood-brain barrier. In wild-type and Naglu-/- mice, CI-MPR expression in brain vasculature is high during the neonatal period but virtually absent by adolescence. In contrast, CI-MPR remains expressed through adolescence in non-affected non-human primate and human brain vasculature. Combined results from IV administration of BMN 250 in Naglu-/- mice and IV and ICV administration in healthy juvenile non-human primates suggest a limitation to therapeutic benefit from IV administration because enzyme distribution is restricted to brain vascular endothelial cells: enzyme does not reach target neuronal cells following IV administration, and pharmacological response following IV administration is likely restricted to clearance of substrate in endothelial cells. In contrast, ICV administration enables central nervous system enzyme replacement with biodistribution to target cells.


Assuntos
Acetilglucosaminidase/administração & dosagem , Acetilglucosaminidase/genética , Barreira Hematoencefálica/química , Fator de Crescimento Insulin-Like II/administração & dosagem , Mucopolissacaridose III/tratamento farmacológico , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Acetilglucosaminidase/uso terapêutico , Administração Intravenosa , Animais , Modelos Animais de Doenças , Terapia de Reposição de Enzimas , Feminino , Infusões Intraventriculares , Fator de Crescimento Insulin-Like II/uso terapêutico , Masculino , Camundongos , Camundongos Transgênicos , Mucopolissacaridose III/genética , Primatas , Proteínas Recombinantes de Fusão/uso terapêutico , Pesquisa Translacional Biomédica
10.
Thromb Res ; 122(4): 533-40, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18539312

RESUMO

Antiplatelet drugs are used to prevent aberrant platelet activation in pathophysiologic conditions such as myocardial infarction and ischemic stroke. The key role that ADP plays in this process has led to the development of antiplatelet drugs that target the P2Y12 receptor. The aim of this study was to characterize the pharmacodynamic (PD) and pharmacokinetic (PK) properties of the novel P2Y12 receptor antagonists, BX 667 and BX 048. BX 667 blocks ADP-induced platelet aggregation in human, dog and rat blood (IC50=97, 317 and 3000 nM respectively). BX 667 had nominal effects on collagen-induced aggregation and weakly inhibited arachidonic acid-induced aggregation. BX 667 has an active metabolite, BX 048, that also potently inhibits ADP-induced aggregation (IC50=290 nM) in human blood. BX 667 was shown to have high oral bioavailability in both dog and rat unlike BX 048. Administration of BX 667 resulted in a rapid and sustained inhibition of platelet aggregation where the extent and duration of platelet inhibition was directly proportional to circulating plasma levels. This report describes the PK/PD properties of BX 667 showing that it has the properties required for a potential antiplatelet therapeutic agent.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Cetoácidos/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Antagonistas do Receptor Purinérgico P2 , Quinolinas/farmacocinética , Receptores Purinérgicos P2/metabolismo , Animais , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Ligantes , Masculino , Modelos Biológicos , Agregação Plaquetária/efeitos dos fármacos , Ligação Proteica , Ratos , Receptores Purinérgicos P2Y12 , Especificidade da Espécie
11.
Thromb Haemost ; 97(1): 45-53, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200770

RESUMO

We have discovered a novel small-molecule (3-phosphinoylpropionic acid) inhibitor of activated thrombin activatable fibrinolysis inhibitor (TAFIa), BX 528, which had an IC (50) of 2 nM in an enzymatic assay and 50 nM in an in-vitro clot lysis assay, with 3,500- to 35,000-fold selectivity against other carboxypeptidases, such as CPN, CPZ and CPD, and 5- and 12-fold selectivity against CPE (CPH) and CPB, respectively. At 10 micro M, BX 528 had no significant activity (<50% inhibition or antagonism) in a panel of 137 enzymes and receptors. It had no effects on blood coagulation and platelet aggregation up to 300 and 10 micro M, respectively. The plasma half-life following intravenous administration was 0.85 hours in rats and 4.5 hours in dogs. No significant metabolism was detected in human, dog or rabbit hepatic microsomes, and no significant inhibition of cytochrome P450 3A4 and 2D6 up to 30 micro M. No cytotoxic or cell proliferative effects were found in three hepatic and renal cell lines up to 300 micro M and no mutagenic activity was seen in the Ames II screen. There were no significant hemodynamic effects in rats and dogs up to 100 and 30 mg/kg with peak plasma drug concentrations of approximately 1,000 and 300 micro M, respectively. In an in-vivo complement activation model in guinea pigs, BX 528 showed minimal inhibition of plasma CPN activity up to 60 mg/kg with peak plasma concentrations up to 250 micro M. Thus, these data demonstrate that BX 528 is a novel, potent, selective and safe TAFIa inhibitor.


Assuntos
Carboxipeptidase B2/antagonistas & inibidores , Inibidores Enzimáticos/farmacocinética , Animais , Coagulação Sanguínea/efeitos dos fármacos , Carboxipeptidases/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Cães , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Cobaias , Meia-Vida , Humanos , Concentração Inibidora 50 , Microssomos Hepáticos/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Propionatos/farmacocinética , Propionatos/farmacologia , Coelhos , Ratos , Especificidade por Substrato
12.
Thromb Haemost ; 97(1): 54-61, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200771

RESUMO

We have discovered a novel small-molecule TAFIa inhibitor, BX 528, which is potent, highly selective against other carboxypeptidases and safe. The present study was to determine if BX 528 can enhance exogenous and endogenous thrombolysis in four different animal models. In the first three models, a thrombus was induced by FeCl (2) (dogs) or laser (rats) injury of the femoral artery, or formed ex vivo and implanted in the jugular vein in rabbits. A low dose of exogenous t-PA was given to induce a low-level thrombolysis on an established thrombus. Co-treatment with BX 528 further enhanced the thrombolytic effects induced by the exogenous t-PA and, thus, reduced thrombosis in all three animal models. In a second rat model, fibrin deposition in the lungs was induced by batroxobin, which was spontaneously resolved in 30 minutes due to the activation of endogenous fibrinolysis. Pre-treatment with lipopolysaccharide (LPS) attenuated this spontaneous fibrinolysis. Co-treatment with 10 mg/kg BX 528 prevented the LPS-induced attenuation of endogenous fibrinolysis. Thus, these studies demonstrated that inhibition of TAFIa by BX 528, our newly discovered small-molecule TAFIa inhibitor, enhanced both the exogenous (induced by a low dose of t-PA) and endogenous (LPS-induced resistance) thrombolysis without increasing the bleeding risk in four different animal models of thrombosis in different species (rat, dog and rabbit) employing different thrombogenic stimuli (FeCl (2) , laser, ex vivo and batroxobin) to induce thrombus formation in different tissues (artery, vein and lung microcirculation).


Assuntos
Carboxipeptidase B2/antagonistas & inibidores , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Trombose/tratamento farmacológico , Animais , Modelos Animais de Doenças , Cães , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Lipopolissacarídeos/farmacologia , Propionatos/farmacologia , Coelhos , Ratos , Ativador de Plasminogênio Tecidual/farmacologia
13.
Thromb Haemost ; 97(5): 847-55, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17479197

RESUMO

Irreversible platelet inhibitors, such as aspirin and clopidogrel, have limited anti-thrombotic efficacy in the clinic due to their bleeding risk. We have developed an orally active reversible P2Y(12) receptor antagonist, BX 667. The aim of this study was to determine if the reversible antagonist BX 667 had a greater therapeutic index than the irreversible P2Y(12) receptor antagonist clopidogrel. Since BX 667 is rapidly converted to its active metabolite BX 048 in rats, we first injected BX 048 intravenously (iv) in a rat arterial venous (A-V) shunt model of thrombosis. BX 048 dose- and concentration-dependently attenuated thrombosis. When administered orally, BX 667 and clopidogrel had similar efficacy, but BX 667 caused less bleeding than clopidogrel. In a rat model of a platelet-rich thrombus induced by vessel injury with FeCl(2), both BX 667 and clopidogrel exhibited higher levels of thrombus inhibition after oral administration compared to their potency in the A-V shunt model. Again, BX 667 caused less bleeding than clopidogrel. In a dog cyclic flow model, iv injection of either BX 667 or clopidogrel dose-dependently reduced thrombus formation with lower bleeding for BX 667 than clopidogrel. Inhibition of thrombosis was highly correlated with inhibition of ADP-induced platelet aggregation in these animal models. In dogs pre-treated with aspirin, BX 667 maintained its wider therapeutic index, measured by inhibition of platelet aggregation over bleeding, compared to the aspirin-clopidogrel combination. These data demonstrate that the reversible P2Y(12) receptor antagonist, BX 667, has a wider therapeutic index than clopidogrel in experimental models of thrombosis.


Assuntos
Proteínas de Membrana/antagonistas & inibidores , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2 , Trombose/prevenção & controle , Animais , Derivação Arteriovenosa Cirúrgica , Lesões das Artérias Carótidas/tratamento farmacológico , Clopidogrel , Modelos Animais de Doenças , Cães , Técnicas In Vitro , Masculino , Estrutura Molecular , Inibidores da Agregação Plaquetária/sangue , Inibidores da Agregação Plaquetária/química , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2 , Receptores Purinérgicos P2Y12 , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia
14.
Mol Ther Methods Clin Dev ; 6: 43-53, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28664165

RESUMO

Sanfilippo syndrome type B (mucopolysaccharidosis IIIB), caused by inherited deficiency of α-N-acetylglucosaminidase (NAGLU), required for lysosomal degradation of heparan sulfate (HS), is a pediatric neurodegenerative disorder with no approved treatment. Intracerebroventricular (ICV) delivery of a modified recombinant NAGLU, consisting of human NAGLU fused with insulin-like growth factor 2 (IGF2) for enhanced lysosomal targeting, was previously shown to result in marked enzyme uptake and clearance of HS storage in the Naglu-/- mouse brain. To further evaluate regional, cell type-specific, and dose-dependent biodistribution of NAGLU-IGF2 (BMN 250) and its effects on biochemical and histological pathology, Naglu-/- mice were treated with 1-100 µg ICV doses (four times over 2 weeks). 1 day after the last dose, BMN 250 (100 µg doses) resulted in above-normal NAGLU activity levels, broad biodistribution, and uptake in all cell types, with NAGLU predominantly localized to neurons in the Naglu-/- mouse brain. This led to complete clearance of disease-specific HS and reduction of secondary lysosomal defects and neuropathology across various brain regions lasting for at least 28 days after the last dose. The substantial brain uptake of NAGLU attainable by this highest ICV dosage was required for nearly complete attenuation of disease-driven storage accumulations and neuropathology throughout the Naglu-/- mouse brain.

15.
Circulation ; 111(17): 2219-26, 2005 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15851596

RESUMO

BACKGROUND: Angiotensin II (Ang II) accelerates atherosclerosis and induces abdominal aortic aneurysm (AAA) in an experimental mouse model. Agonism of a G protein-coupled receptor by Ang II activates Rho-kinase and other signaling pathways and results in activation of proteolysis and apoptosis. Enhanced proteolysis and smooth muscle cell apoptosis are important mechanisms associated with AAA. In this study, we tested the hypothesis that fasudil, a Rho-kinase inhibitor, could attenuate Ang II-induced AAA formation by inhibiting vascular wall apoptosis and extracellular matrix proteolysis. METHODS AND RESULTS: Six-month-old apolipoprotein E-deficient mice were infused with Ang II (1.44 mg x kg(-1) x d(-1)) for 1 month. Animals were randomly assigned to treatment with fasudil (136 or 213 mg x kg(-1) x d(-1) in drinking water) or tap water. Ang II infusion induced AAA formation in 75% of the mice, which was accompanied by an increase in proteolysis detected by zymographic analysis and quantified by active matrix metalloproteinase-2 activity, as well as apoptosis detected by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and quantified by both caspase-3 activity and histone-associated DNA fragmentation. The level of DNA fragmentation in the suprarenal aorta correlated with AAA diameter. Ang II also increased atherosclerotic lesion area and blood pressure. Fasudil treatment resulted in a dose-dependent reduction in both the incidence and severity of AAA. At the higher dose, fasudil decreased AAA by 45% while significantly inhibiting both apoptosis and proteolysis, without affecting atherosclerosis or blood pressure. CONCLUSIONS: These data demonstrate that inhibition of Rho-kinase by fasudil attenuated Ang II-induced AAA through inhibition of both apoptosis and proteolysis pathways.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Angiotensina II/farmacologia , Aneurisma da Aorta Abdominal/tratamento farmacológico , Apolipoproteínas E/deficiência , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Apoptose , Endotélio Vascular/citologia , Matriz Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Inibidores de Proteases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinases Associadas a rho
16.
Thromb Haemost ; 96(3): 317-24, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16953273

RESUMO

Tissue factor (TF) exposure is a potent pro-thrombotic trigger that initiates activation of the coagulation cascade, while thrombomodulin (TM) is a potent anticoagulant protein that limits the extent of activation. Both TF neutralizing antibodies and soluble TM (sTM) are effective anticoagulants. We have developed a novel anticoagulant fusion protein, Ab(TF)-TM, by fusing a TFneutralizing single-chain antibody, Ab(TF), to an active fragment of TM. Ab(TF)-TM is a novel anticoagulant targeting to sites of TF exposure with a dual mechanism of action. The Ab(TF) portion of the molecule inhibitsTF/factorVIIa mediated activation of FIX and FX, and the TM portion of the molecule acts as a cofactor for activation of protein C. In-vitro coagulation assays show that Ab(TF)-TM more potently inhibits TF-initiated coagulation (prothrombin time) than can its individual components, Ab(TF) (20-fold) and sTM (80-fold) alone, or in combination (10-fold). In contrast, the potency of Ab(TF)-TM in the activated partial thromboplastin and thrombin clotting time assays was similar to sTM alone. In a rat model of disseminated intravascular coagulation (DIC), intravenous injection of a human TF-containing thromboplastin reagent (0.5 ml/kg) resulted in an immediate death in approximately 60% of the animals and a clinical score of approximately 2.5. Pre-injection of Ab(TF)-TM or Ab(TF) and sTM, given alone or in combination, showed dose-dependent efficacy. At a dose of 0.7 nmol/kg, Ab(TF)-TM completely prevented death and reduced clinical scores by 79%, while neitherAb(TF) nor sTM, given alone or in combination, showed significant therapeutic effects. Calculated effective doses that reduced mortality by 50% relative to that in the control group (ED(50), nmol/kg) were 0.21 for Ab(TF)-TM, 3.2 for an equimolar mixture of Ab(TF) and sTM, 4.3 for sTM and 20 for Ab(TF). Thus, Ab(TF)-TM presented 10- to 100-fold enhancement of the anticoagulant potency, relative to the ED(50) in Ab(TF) and sTM given either alone or in combination, in a rat DIC model.


Assuntos
Anticoagulantes/farmacologia , Trombomodulina/sangue , Tromboplastina/metabolismo , Animais , Anticoagulantes/química , Coagulação Sanguínea , Dimerização , Relação Dose-Resposta a Droga , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Tromboembolia , Trombomodulina/química , Tromboplastina/imunologia , Fatores de Tempo
17.
J Alzheimers Dis ; 52(1): 223-42, 2016 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-27060954

RESUMO

Recent studies have shown that inoculation of susceptible mice with amyloid-ß (Aß) peptides accelerates Aß deposition in the brain, supporting the idea that Aß may be self-amplifying; however, the exact mechanism is not understood. Here we provide evidence that Aß may self-amplify, in part, by inhibiting α-secretase ADAM10 (a disintegrin and metalloprotease) cleavage of full-length Aß precursor protein (FL AßPP) and therefore allow greater ß-secretase processing, and that Aß itself is a substrate for ADAM10. Exposure of primary neuronal cultures from PDAßPP mice to exogenous rat Aß1- 40 resulted in increased de novo human Aß1-42 production and exposure of cells to Aß decreased production of ADAM10 cleavage product soluble AßPPα (sAßPPα). In a cell-free assay, Aß decreased ADAM10 cleavage of the chimeric substrate MBP-AßPPC125 and Aß itself was apparently cleaved by the enzyme. The axonal guidance and trophic factor netrin-1, however, reduced the Aß1- 40-induced Aß1-42 increase, increased sAßPPα, and reversed the Aß-induced sAßPPα decrease in vitro. In vivo, induction of netrin-1 expression in PDAßPPSwe/Ind transgenic mice resulted in reductions in both Aß1-42 and Aß1- 40, and ICV delivery of netrin-1 to PDAßPPSwe/Ind mice increased sAßPPα, decreased Aß, and improved working memory. Finally, to support further study of netrin-1's potential as a therapeutic for Alzheimer's disease, pilot gene therapy studies were performed and a netrin mimetic peptide synthesized and tested that, like netrin, can increase sAßPPα and decrease Aß1-42in vitro. Taken together, these data provide mechanistic insights into Aß self-amplification and the ability of netrin-1 to disrupt it.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/metabolismo , Terapia Genética/métodos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína ADAM10/metabolismo , Doença de Alzheimer/psicologia , Animais , Biomimética , Linhagem Celular Tumoral , Cognição/fisiologia , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Memória de Curto Prazo/fisiologia , Camundongos Transgênicos , Netrina-1 , Projetos Piloto , Ratos , Reconhecimento Psicológico/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Eur J Pharmacol ; 512(2-3): 215-22, 2005 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-15840407

RESUMO

Recent evidence indicates that the GTPase activated Rho/Rho-kinase pathway contributes angiotensin II-induced cardiac hypertrophy and vascular remodeling. We tested this hypothesis in vivo by determining the effects of fasudil, a Rho-kinase inhibitor, on angiotensin II-induced cardiac hypertrophy, coronary vascular remodeling, and ventricular dysfunction. Six-month-old apolipoprotein E deficient (apoE-KO) mice were subcutaneously infused with angiotensin II (1.44 mg/kg/day) using an osmotic mini-pump. Mice were randomly assigned to either vehicle or fasudil (136 or 213 mg/kg/day in drinking water) group. Infusion of angiotensin II for 4 weeks resulted in cardiac enlargement, myocyte hypertrophy, and myocardial interstitial and coronary artery perivascular fibrosis. These changes were accompanied by reduced aortic flow velocity and acceleration rate. Cardiac gene expression levels of atrial natriuretic peptide (ANP) and collagen type III detected by real-time reverse transcriptase polymerase chain reaction were significantly increased in angiotensin II-infused mice. Treatment with fasudil dose-dependently attenuated angiotensin II-induced cardiac hypertrophy, prevented perivascular fibrosis, blunted the increase in ANP and collagen type III expression, and improved cardiac function, without changing blood pressure. These data are consistent with a role for Rho-kinase activation in angiotensin II-induced cardiac remodeling and vascular wall fibrosis.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Angiotensina II/farmacologia , Apolipoproteínas E/genética , Cardiomegalia/prevenção & controle , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Apolipoproteínas E/metabolismo , Fator Natriurético Atrial/genética , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Colágeno Tipo III/genética , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Relação Dose-Resposta a Droga , Fibrose/prevenção & controle , Expressão Gênica/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Quinases Associadas a rho
19.
Ultrasound Med Biol ; 31(6): 745-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15936490

RESUMO

Mouse models of abdominal aortic aneurysm (AAA) have been commonly used in many laboratories for studying molecular mechanisms of AAA formation and development, as well as for testing novel therapeutic agents in the treatment of AAA. However, because of the small size of the animal, the quantification and characterization of AAA development and progress is difficult, time-consuming and requires the sacrifice of the experimental animals. We report here a noninvasive method to detect and measure AAA in mice using a high-frequency ultrasound (US) imaging system specifically designed for microimaging of the mice (Vevo 660; VisualSonics, Toronto, ONT, Canada). A total of 21 male apolipoprotein-E-deficient mice were chronically infused with angiotensin II (1.44 mg/kg daily) for 28 days to induce AAA formation. A 2-D echo image of the abdominal aorta was acquired at longitudinal and transverse planes, followed immediately by post mortem dissection of the abdominal aorta for direct measurements. The US images clearly showed a bulge-like expansion localized specifically in the suprarenal region of the abdominal aorta, with a shape strikingly similar to that of the aorta dissected post mortem. In addition, the US images can also provide measurements of the luminal diameter and wall thickness of the abdominal aorta. The average dimensions of the abdominal aorta were not significantly different between the US and post mortem measurements, nor between the transverse and longitudinal US images. The different types of the measurements are also highly correlated with each other, with a linear correlation (r) between 0.7 and 0.9. Thus, we have established and validated a novel application to noninvasively measure AAA development and progress in a mouse model using a high-frequency US imaging system that has the advantages of low cost, rapid imaging speed, reproducibility and high resolution, and makes repeated monitoring of the progress of AAA development over a time-course possible.


Assuntos
Aorta Abdominal/diagnóstico por imagem , Aneurisma da Aorta Abdominal/diagnóstico por imagem , Processamento de Sinais Assistido por Computador , Animais , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/patologia , Dissecação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Sensibilidade e Especificidade , Ultrassonografia
20.
Thromb Haemost ; 92(5): 956-65, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15543321

RESUMO

During thrombosis, P-selectin is expressed on the surface of activated endothelial cells and platelets. We hypothesized that targeting a plasminogen activator (PA) to P-selectin would enhance local thrombolysis and reduce bleeding risk. Previously, a urokinase (uPA)/anti-P-selectin antibody (HuSZ51) fusion protein was shown to increase fibrinolysis in a hamster pulmonary embolism model. To explore the therapeutic potential of this targeting strategy, we fused the fibrin-selective Desmodus rotundus salivary PA alpha1 (dsPA alpha 1) to HuSZ51 and compared the fibrinolytic activity of P-selectin-targeted dsPA alpha 1 (HuSZ51-dsPA alpha 1) to unmodified dsPA alpha 1 in vitro and in vivo. HuSZ51-dsPA alpha 1 and dsPA alpha 1 were expressed in CHO cells and purified to homogeneity by affinity chromatography. HuSZ51-dsPA alpha 1 bound to thrombin-activated human and dog platelets with comparable affinities to that of parental antibody SZ51. The fusion protein retained the catalytic activities of dsPA alpha 1 in chromogenic and clot lysis assays, indicating that dsPA alpha 1 is fully functional when fused to HuSZ51. Compared to dsPA alpha 1, HuSZ51-dsPA alpha 1 had similar thrombolytic efficacy in a rat pulmonary embolism model and anti-thrombotic potency in a dog model of femoral artery thrombosis. However, HuSZ51-dsPA alpha 1 was less effective in lysis of preexisting arterial thrombi in the dog model. The reduced arterial thrombolysis was not due to the pharmacokinetic properties of HuSZ51-dsPA alpha 1 because antigen level and amidolytic activity were higher in plasma from HuSZ51-dsPA alpha 1-treated groups than corresponding dsPA alpha 1-treated groups. These data indicate that the thrombolytic efficacy of HuSZ51-dsPA alpha 1 varied dependent on the physical composition of thrombi. The lack of stimulation by fibrin in arterial thrombi may contribute to the attenuated thrombolytic efficacy of HuSZ51-dsPA alpha 1 in the dog model.


Assuntos
Plaquetas/efeitos dos fármacos , Fibrina/efeitos dos fármacos , Imunoconjugados/uso terapêutico , Selectina-P/imunologia , Ativadores de Plasminogênio/administração & dosagem , Terapia Trombolítica/métodos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/uso terapêutico , Plaquetas/química , Plaquetas/fisiologia , Modelos Animais de Doenças , Cães , Fibrinolíticos/administração & dosagem , Fibrinolíticos/farmacocinética , Fibrinolíticos/farmacologia , Engenharia Genética , Humanos , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Ativadores de Plasminogênio/genética , Ativadores de Plasminogênio/uso terapêutico , Agregação Plaquetária , Embolia Pulmonar/tratamento farmacológico , Ratos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Trombina , Trombose/tratamento farmacológico , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA