Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Eur J Pharmacol ; 909: 174439, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34425100

RESUMO

Phillyrin, a natural plant extract, has significant antioxidant and anti-apoptotic effects. However, its effect on intracerebral hemorrhage (ICH) remains unclear. In this study, we investigated a potential role for phillyrin in the regulation of the oxidative stress and apoptosis induced by ICH. A model of ICH was induced by collagenase IV (0.2 U in 1 µl sterile normal saline) in male C57BL/6J (B6) mice and different doses of phillyrin (5, 15, or 30 mg/kg) were intraperitoneally (i.p.) injected at 30 min, 6 h, and 22 h after modeling. We found that phillyrin significantly reduced neural function and lesion volume, improved injury of white and grey matter around the lesion, decreased apoptosis and oxidative stress, increased the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase 1(HO-1), NADPH quinone oxidoreductase 1 (NQO1) and Superoxide Dismutase-1(SOD-1) in vitro and in vivo, and protected neurons from the stimulation of hemin by promoting Nrf2 nuclear translocation. Treatment with ML385 (Nrf2 inhibitor) completely reversed the protective effects of phillyrin in vivo after ICH injury. Based on our findings, we conclude that phillyrin treatment alleviates ICH injury-induced apoptosis and oxidative stress via activation of the Nrf2 signaling pathway, highlighting a potential role for phillyrin as an ICH therapeutic.


Assuntos
Hemorragia Cerebral/tratamento farmacológico , Glucosídeos/farmacologia , Fator 2 Relacionado a NF-E2/agonistas , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Hemorragia Cerebral/imunologia , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Glucosídeos/uso terapêutico , Humanos , Imidazolidinas/administração & dosagem , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/imunologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Compostos de Espiro/administração & dosagem
2.
J Med Chem ; 63(15): 8046-8058, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32407115

RESUMO

Respiratory syncytial virus (RSV) is a seasonal virus that infects the lungs and airways of 64 million children and adults every year. It is a major cause of acute lower respiratory tract infection and is associated with significant morbidity and mortality. Despite the large medical and economic burden, treatment options for RSV-associated bronchiolitis and pneumonia are limited and mainly consist of supportive care. This publication covers the medicinal chemistry efforts resulting in the identification of JNJ-53718678, an orally bioavailable RSV inhibitor that was shown to be efficacious in a phase 2a challenge study in healthy adult subjects and that is currently being evaluated in hospitalized infants and adults. Cocrystal structures of several new derivatives helped in rationalizing some of the structure-activity relationship (SAR) trends observed.


Assuntos
Antivirais/química , Descoberta de Drogas/métodos , Imidazolidinas/química , Indóis/química , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/química , Administração Oral , Antivirais/administração & dosagem , Cristalografia por Raios X/métodos , Células HeLa , Humanos , Imidazolidinas/administração & dosagem , Indóis/administração & dosagem , Estrutura Secundária de Proteína , Vírus Sincicial Respiratório Humano/fisiologia , Inibidores de Proteínas Virais de Fusão/administração & dosagem
3.
J Med Chem ; 63(10): 5089-5099, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32022560

RESUMO

We have previously shown that the oral administration of the small molecule hPTHR1 agonist PCO371 and its lead compound, 1 (CH5447240) results in PTH-like calcemic and hypophostemic activity in thyroparathyroidectomized rats. However, 1 was converted to a reactive metabolite in a human liver microsome assay. In this article, we report on the modification path that led to an enhancement of PTHR1 agonistic activity and reduction in the formation of a reactive metabolite to result in a potent, selective, and orally active PTHR1 agonist 1-(3,5-dimethyl-4-(2-((4-oxo-2-(4-(trifluoromethoxy)phenyl)-1,3,8-triazaspiro[4.5]dec-1-en-8-yl)sulfonyl)ethyl)phenyl)-5,5-dimethylimidazolidine-2,4-dione (PCO371, 16c). This compound is currently being evaluated in a phase 1 clinical study for the treatment of hypoparathyroidism.


Assuntos
Imidazolidinas/administração & dosagem , Imidazolidinas/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/agonistas , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Compostos de Espiro/administração & dosagem , Compostos de Espiro/metabolismo , Administração Oral , Animais , Feminino , Humanos , Hipoparatireoidismo/tratamento farmacológico , Hipoparatireoidismo/metabolismo , Imidazolidinas/química , Células LLC-PK1 , Ratos , Ratos Sprague-Dawley , Compostos de Espiro/química , Suínos
4.
Bioorg Med Chem Lett ; 29(16): 2387-2392, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31208765

RESUMO

The aim of this study was to design and synthesize two series of N-Mannich bases with imidazolidine-2,4-dione core as a potential anticonvulsant with reduced toxicity and broad antiseizure activity. Preliminary screening revealed that the majority of synthesized compounds were effective in the maximal electroshock seizure (MES) and/or subcutaneous pentylenetetrazole (scPTZ) test. The most active in vivo compound, 18 (3-((4-methylpiperazin-1-yl)methyl)-5,5-diphenylimidazolidine-2,4-dione), exhibited an ED50 value comparable to that of phenytoin in the MES test (38.5 mg/kg vs 28.1 mg/kg), and more importantly, it showed four times higher potency than phenytoin in the 6 Hz test (12.2 mg/kg vs > 60 mg/kg). Additionally, 18 exhibited antiallodynic properties in the von Frey test in neuropathic (oxaliplatin-treated) mice. Compound 18 also demonstrated a broader spectrum of anticonvulsant activity than phenytoin and showed statistically significant antinociceptive properties in selected models of chronic pain.


Assuntos
Anticonvulsivantes/uso terapêutico , Imidazolidinas/uso terapêutico , Bases de Mannich/uso terapêutico , Dor/tratamento farmacológico , Convulsões/tratamento farmacológico , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/síntese química , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Imidazolidinas/administração & dosagem , Imidazolidinas/síntese química , Bases de Mannich/administração & dosagem , Bases de Mannich/síntese química , Camundongos , Estrutura Molecular , Oxaliplatina , Dor/induzido quimicamente , Ratos , Convulsões/induzido quimicamente , Relação Estrutura-Atividade
5.
Acta Neuropathol Commun ; 7(1): 54, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30961664

RESUMO

Recently, moderate prenatal alcohol exposure (PAE) was shown to be a risk factor for peripheral neuropathy following minor nerve injury. This effect coincides with elevated spinal cord astrocyte activation and ex vivo immune cell reactivity assessed by proinflammatory cytokine interleukin (IL) -1ß protein expression. Additionally, the ß2-integrin adhesion molecule, lymphocyte function-associated antigen-1 (LFA-1), a factor that influences the expression of the proinflammatory/anti-inflammatory cytokine network is upregulated. Here, we examine whether PAE increases the proinflammatory immune environment at specific anatomical sites critical in the pain pathway of chronic sciatic neuropathy; the damaged sciatic nerve (SCN), the dorsal root ganglia (DRG), and the spinal cord. Additionally, we examine whether inhibiting LFA-1 or IL-1ß actions in the spinal cord (intrathecal; i.t., route) could alleviate chronic neuropathic pain and reduce spinal and DRG glial activation markers, proinflammatory cytokines, and elevate anti-inflammatory cytokines. Results show that blocking the actions of spinal LFA-1 using BIRT-377 abolishes allodynia in PAE rats with sciatic neuropathy (CCI) of a 10 or 28-day duration. This effect is observed (utilizing immunohistochemistry; IHC, with microscopy analysis and protein quantification) in parallel with reduced spinal glial activation, IL-1ß and TNFα expression. DRG from PAE rats with neuropathy reveal significant increases in satellite glial activation and IL-1ß, while IL-10 immunoreactivity is reduced by half in PAE rats under basal and neuropathic conditions. Further, blocking spinal IL-1ß with i.t. IL-1RA transiently abolishes allodynia in PAE rats, suggesting that IL-1ß is in part, necessary for the susceptibility of adult-onset peripheral neuropathy caused by PAE. Chemokine mRNA analyses from SCN, DRG and spinal cord reveal that increased CCL2 occurs following CCI injury regardless of PAE and BIRT-377 treatment. These data demonstrate that PAE creates dysregulated proinflammatory IL-1ß and TNFα /IL-10 responses to minor injury in the sciatic-DRG-spinal pain pathway. PAE creates a risk for developing peripheral neuropathies, and LFA-1 may be a novel therapeutic target for controlling dysregulated neuroimmune actions as a consequence of PAE.


Assuntos
Antígeno-1 Associado à Função Linfocitária/imunologia , Neuralgia/imunologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Medula Espinal/imunologia , Animais , Astrócitos/imunologia , Feminino , Imidazolidinas/administração & dosagem , Interleucina-10/imunologia , Interleucina-1beta/imunologia , Masculino , Microglia/imunologia , Mielite/imunologia , Gravidez , Ratos Long-Evans
6.
Curr Clin Pharmacol ; 14(2): 108-115, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30648519

RESUMO

BACKGROUND: Prostate cancer is the sixth leading cause of death, among all cancer deaths By 2030, this burden is expected to increase with 1.7 million new cases and 499,000 new deaths. We aimed to evaluate the efficacy and safety of Nilutamide in metastatic prostate cancer (mPCa) patients who underwent orchiectomy. METHODS: A comprehensive search was conducted in the Medline/PubMed and Cochrane Library. References from included studies and studies from clinicaltrials.gov were explored without language and date restrictions. We included only randomized controlled trials, comparing the safety and efficacy of Nilutamide in Metastatic Prostate Cancer (mPCa) patients who underwent orchiectomy with placebo. The outcomes of concerns were survival and the response of drug and safety.. Quality of the included studies was assessed using the Cochrane Risk of Bias Tool. Two authors were independently involved in the study selection, data extraction and quality assessment. Disagreements between the two reviewers were resolved by consulting a third reviewer. RESULTS: A total of five out of 244 studies were included in meta-analysis involving1637 participants. Nilutamide group showed improved response rate (RR=1.77, 95%CI 1.46-2.14, p<0.00001), disease progression (RR=0.59, 95%CI 0.47-0.73, p<0.00001), complete response (RR=2.13, 95%CI 1.40-3.23, p=0.003) and clinical benefit (RR=1.23, 95%CI 1.13-1.34, p<0.00001) when compared to placebo; however, stable disease favored the control group (RR=0.80, 95%CI 0.68-0.94, p=0.007). In addition, patients on Nilutamide showed prolonged progression-free survival and overall survival. Nausea and vomiting were the most common adverse events reported in Nilutamide group. CONCLUSION: Evidence suggests that patients with mPCa who underwent orchiectomy receiving Nilutamide showed significant improvement in progression-free survival and overall survival response rate and clinical benefits in comparison with the placebo group.


Assuntos
Antineoplásicos/administração & dosagem , Imidazolidinas/administração & dosagem , Neoplasias da Próstata/terapia , Antineoplásicos/efeitos adversos , Progressão da Doença , Intervalo Livre de Doença , Humanos , Imidazolidinas/efeitos adversos , Masculino , Metástase Neoplásica , Orquiectomia , Neoplasias da Próstata/patologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Taxa de Sobrevida
7.
J Infect Dis ; 218(5): 748-756, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29684148

RESUMO

Background: Respiratory syncytial virus (RSV) disease has no effective treatment. JNJ-53718678 is a fusion inhibitor with selective activity against RSV. Methods: After confirmation of RSV infection or 5 days after inoculation with RSV, participants (n = 69) were randomized to JNJ-53718678 75 mg (n = 15), 200 mg (n = 17), 500 mg (n = 18), or placebo (n = 17) orally once daily for 7 days. Antiviral effects were evaluated by assessing RSV RNA viral load (VL) area under the curve (AUC) from baseline (before the first dose) until discharge, time-to-peak VL, duration of viral shedding, clinical symptoms, and quantity of nasal secretions. Results: Mean VL AUC was lower for individuals treated with different doses of JNJ-53718678 versus placebo (203.8-253.8 vs 432.8 log10 PFUe.hour/mL). Also, mean peak VL, time to peak VL, duration of viral shedding, mean overall symptom score, and nasal secretion weight were lower in each JNJ-53718678-treated group versus placebo. No clear exposure-response relationship was observed. Three participants discontinued due to treatment-emergent adverse events of grade 2 and 1 electrocardiogram change (JNJ-53718678 75 mg and 200 mg, respectively) and grade 2 urticaria (placebo). Conclusions: JNJ-53718678 at all 3 doses substantially reduced VL and clinical disease severity, thus establishing clinical proof of concept and the compound's potential as a novel RSV treatment. Clinical trials registration: ClinicalTrials.gov: NCT02387606; EudraCT number: 2014-005041-41.


Assuntos
Antivirais/administração & dosagem , Imidazolidinas/administração & dosagem , Indóis/administração & dosagem , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Administração Oral , Adolescente , Adulto , Antivirais/farmacologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Voluntários Saudáveis , Humanos , Imidazolidinas/farmacologia , Indóis/farmacologia , Masculino , Pessoa de Meia-Idade , Placebos/administração & dosagem , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/isolamento & purificação , Resultado do Tratamento , Carga Viral , Eliminação de Partículas Virais , Adulto Jovem
8.
Sci Rep ; 7(1): 3182, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28600556

RESUMO

Anthracycline drugs such as doxorubicin (DOX) and daunorubicin remain some of the most active wide-spectrum and cost-effective drugs in cancer therapy. However, colorectal cancer (CRC) cells are inherently resistant to anthracyclines which at higher doses cause cardiotoxicity. Our recent studies indicate that aldose reductase (AR) inhibitors such as fidarestat inhibit CRC growth in vitro and in vivo. Here, we show that treatment of CRC cells with fidarestat increases the efficacy of DOX-induced death in HT-29 and SW480 cells and in nude mice xenografts. AR inhibition also results in higher intracellular accumulation of DOX and decreases the expression of drug transporter proteins MDR1, MRP1, and ABCG2. Further, fidarestat also inhibits DOX-induced increase in troponin-I and various inflammatory markers in the serum and heart and restores cardiac function in mice. These results suggest that fidarestat could be used as adjuvant therapy to enhance DOX sensitivity of CRC cells and to reduce DOX-associated cardiotoxicity.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Cardiotoxicidade/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Doxorrubicina/administração & dosagem , Imidazolidinas/administração & dosagem , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Aldeído Redutase/genética , Animais , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Doxorrubicina/efeitos adversos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas de Neoplasias/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Zoo Wildl Med ; 48(4): 1077-1080, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29297825

RESUMO

Eleven cases of dilated cardiomyopathy have been diagnosed and treated in captive Livingstone fruit bats ( Pteropus livingstonii) in the United Kingdom over the past 7 yr. All but one case received treatment with a diuretic plus an angiotensin-converting enzyme inhibitor (ACEI), and, or pimobendan. One case is still under treatment with pimobendan alone, following diagnosis before onset of clinical signs. Diuretic treatment consisted of furosemide at a dose rate of 0.5-5 mg/kg, one to three times daily, and, or spironolactone at a dose rate of 1-4 mg/kg, once or twice daily. When used, the ACEI imidapril was given at a dose rate of 0.24-0.38 mg/kg q 24 hr, and pimobendan at a dose rate of 0.2-0.5 mg/kg bid. This report is intended to provide anyone seeking to medically manage heart failure in Pteropus species, particularly P. livingstonii, with a review of drugs and doses that have been used.


Assuntos
Cardiomiopatia Dilatada/veterinária , Quirópteros , Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Animais de Zoológico , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiotônicos/uso terapêutico , Diuréticos/administração & dosagem , Diuréticos/uso terapêutico , Feminino , Furosemida/administração & dosagem , Furosemida/uso terapêutico , Imidazolidinas/administração & dosagem , Imidazolidinas/uso terapêutico , Masculino , Piridazinas/administração & dosagem , Piridazinas/uso terapêutico , Espironolactona/administração & dosagem , Espironolactona/uso terapêutico
11.
Biomed Res Int ; 2016: 9172157, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27088094

RESUMO

Kidney fibrosis is the final common pathway of progressive kidney diseases including diabetic nephropathy. Here, we report that the endogenous antifibrotic peptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), the substrate of angiotensin-converting enzyme (ACE), is an orally available peptide drug used to cure kidney fibrosis in diabetic mice. We utilized two mouse models of diabetic nephropathy, streptozotocin- (STZ-) induced type 1 diabetic CD-1 mice and type 2 diabetic nephropathy model db/db mice. Intervention with the ACE inhibitor imidapril, oral AcSDKP, or imidapril + oral AcSDKP combination therapy increased urine AcSDKP levels. AcSDKP levels were significantly higher in the combination group compared to those of the other groups. AcSDKP oral administration, either AcSDKP alone or in addition to imidapril, ameliorated glomerulosclerosis and tubulointerstitial fibrosis. Plasma cystatin C levels were higher in both models, at euthanasia, and were restored by all the treatment groups. The levels of antifibrotic miRs, such as miR-29 or let-7, were suppressed in the kidneys of both models; all treatments, especially the combination of imidapril + oral AcSDKP, restored the antifibrotic miR levels to a normal value or even higher. AcSDKP may be an oral antifibrotic peptide drug that would be relevant to combating fibroproliferative kidney diseases such as diabetic nephropathy.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Nefropatias Diabéticas/tratamento farmacológico , Oligopeptídeos/administração & dosagem , Administração Oral , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/patologia , Humanos , Imidazolidinas/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Peptidil Dipeptidase A/efeitos dos fármacos , Peptidil Dipeptidase A/metabolismo
12.
Prog Retin Eye Res ; 54: 1-29, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27102270

RESUMO

Diabetes mellitus (DM) is a major health problem with devastating effects on ocular health in both industrialized and developing countries. The control of hyperglycemia is critical to minimizing the impact of DM on ocular tissues because inadequate glycemic control leads to ocular tissue changes that range from a temporary blurring of vision to permanent vision loss. The biochemical mechanisms that promote the development of diabetic complications have been extensively studied. As a result, a number of prominent biochemical pathways have been identified. Among these, the two-step sorbitol pathway has been the most extensively investigated; nevertheless, it remains controversial. To date, long-term pharmacological studies in animal models of diabetes have demonstrated that the onset and development of ocular complications that include keratopathy, retinopathy and cataract can be ameliorated by the control of excess metabolic flux through aldose reductase (AR). Clinically the alleles of AR have been linked to the rapidity of onset and severity of diabetic ocular complications in diabetic patient populations around the globe. In spite of these promising preclinical and human genetic rationales, several clinical trials of varying durations with structurally diverse aldose reductase inhibitors (ARIs) have shown limited success or failure in preventing or arresting diabetic retinopathy. Despite these clinical setbacks, topical ARI Kinostat(®) promises to find a home in clinical veterinary ophthalmology where its anticipated approval by the FDA will present an alternative treatment paradigm to cataract surgery in diabetic dogs. Here, we critically review the role of AR in diabetes mellitus-linked ocular disease and highlight the development of Kinostat(®) for cataract prevention in diabetic dogs. In addition to the veterinary market, we speculate that with further safety and efficacy studies in humans, Kinostat(®) or a closely related product could have a future role in treating diabetic keratopathy.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Catarata/tratamento farmacológico , Doenças da Córnea/tratamento farmacológico , Complicações do Diabetes , Retinopatia Diabética/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Imidazolidinas/uso terapêutico , Administração Tópica , Animais , Diabetes Mellitus/metabolismo , Inibidores Enzimáticos/administração & dosagem , Humanos , Imidazolidinas/administração & dosagem , Soluções Oftálmicas
13.
Expert Opin Ther Pat ; 26(6): 731-5, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26967920

RESUMO

Diabetes mellitus is a major threat to global public health that requires long-term medical attention. In view of the potentially devastating effects of diabetes on ocular health, it highlights the urgent need of therapeutic drugs for the prevention and treatment of the diabetic complications. The patent described in this evaluation (WO2015026380A1) claimed a topical composition for treating diabetic cataracts both in animals and human beings. The composition containing a therapeutic amount of the 2R-methyl sorbinil, one of aldose reductase inhibitors, delivered to the dog's eye can exert a preventive, inhibitory, or prophylactic effect on diabetic cataracts in a statistically significant portion of the population being studied. Thus methods and strategies using new formulations of known inhibitors are promising for future use in the treatment of diabetic complications.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Catarata/tratamento farmacológico , Complicações do Diabetes/tratamento farmacológico , Administração Oftálmica , Animais , Catarata/etiologia , Catarata/veterinária , Complicações do Diabetes/patologia , Complicações do Diabetes/veterinária , Cães , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Imidazolidinas/administração & dosagem , Imidazolidinas/farmacologia , Imidazolidinas/uso terapêutico , Patentes como Assunto
14.
Int Immunopharmacol ; 24(2): 153-158, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25499727

RESUMO

Liver plays a central role in xenobiotics metabolism, thus affecting the in vivo disposition and therapeutic effects of drugs. Carboxylesterases (CESs), with the main isoforms CES1 and CES2, are important in the metabolism of ester-type prodrugs. However, influences of immunological liver injury on the activity of CES remain undefined. In the present study, we demonstrated treatment with lipopolysaccharide (LPS) suppressed the activities of CES1 and CES2. The decreased activities of CES1 and CES2 were preliminarily assessed by the hydrolysis assay for their common substrate p-nitrophenyl acetate (PNPA) with rat hepatic microsomal enzyme. Subsequently, RT-PCR results showed that the levels of CES1 mRNA and mRNA of CES2 (AB010635) and CES2 (AY034877) in the model group were significantly lower than those of the normal control group (P<0.05). Western blot results showed that the expressions of CES1 and CES2 proteins were decreased (P<0.05). To further clarify the effects of LPS on the metabolic activities of CESs, pharmacokinetic studies were performed in rats by utilizing imidapril and irinotecan (CPT-11) as the specific substrates for CES1 and CES2, respectively. After treatment with LPS, AUC0-∞ and Cmax of imidaprilat were decreased from 2084.86±340.66ng·h(-1)·mL(-1) and 234.66±68.85ng·mL(-1) to 983.87±315.34ng·h(-1)·mL(-1) and 113.1±19.69ng·mL(-1) (P<0.05), respectively. Moreover, AUC0-∞ and Cmax of SN-38 were decreased from 8100±918.6ng·h(-1)·mL(-1) and 144.67±20.28ng·mL(-1) to 3270±500.5ng·h(-1)·mL(-1) and 56.19±10.38ng·mL(-1) (P<0.05), respectively. In summary, immunological liver injury remarkably attenuated the expressions and metabolic activities of CES1 and CES2, which may be associated with the regulatory effects of cytokines under inflammation.


Assuntos
Camptotecina/análogos & derivados , Hidrolases de Éster Carboxílico/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Imidazolidinas/farmacocinética , Fígado/metabolismo , Animais , Camptotecina/administração & dosagem , Camptotecina/farmacocinética , Hidrolases de Éster Carboxílico/genética , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Regulação para Baixo , Imidazolidinas/administração & dosagem , Irinotecano , Lipopolissacarídeos/imunologia , Fígado/efeitos dos fármacos , Masculino , Mycobacterium bovis/imunologia , Pró-Fármacos/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato
15.
Tumour Biol ; 35(1): 323-32, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23959470

RESUMO

Rosiglitazone (RGZ) and imidapril improve cancer cachexia via different mechanisms. Therefore, we hypothesized that combination therapy of RGZ+imidapril would further attenuate cancer cachexia in vivo. After injection with colon-26 adenocarcinoma for 9 days, BALB/c mice were randomly divided into the following four treatment groups for 7 days (n = 8 per group): (1) placebo, (2) RGZ, (3) imidapril, and (4) RGZ+imidapril. Eight healthy control animals were also assessed. Body weight, tumor volume, gastrocnemius muscle and epididymal adipose mass, serum metabolic markers and cytokines, and the expression of nuclear factor-κB and two E3 ubiquitin ligases, atrogin-1 and MuRF-1, were measured. From days 14 to 16, all treatments significantly reduced tumor volume (P < 0.05). From days 10 to 16, improvements in the tumor-free body weight were observed in the RGZ and RGZ+imidapril groups. In addition, significant improvements in both gastrocnemius muscle and epididymal adipose mass were observed in all treatment groups (all, P < 0.05). Furthermore, all treatments significantly increased tumor necrosis factor alpha levels as compared to those observed in the healthy control animals (P < 0.001). Insulin levels significantly increased in the placebo group as compared to those in the healthy control group (P < 0.05), which were reduced in all the treatment groups (P < 0.05). Finally, whereas all treatments significantly reduced atrogin-1 levels as compared to the placebo group (all, P < 0.05), significant reductions in MuRF-1 levels were only observed in the RGZ and RGZ+imidapril groups (both, P < 0.05). Thus, all three treatments reduce tumor growth and alleviate cancer cachexia; however, synergistic effects of RGZ+imidapril combination therapy were not observed.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Caquexia/etiologia , Caquexia/patologia , Imidazolidinas/farmacologia , Músculos/efeitos dos fármacos , Neoplasias/complicações , Tiazolidinedionas/farmacologia , Tecido Adiposo/patologia , Animais , Biomarcadores/sangue , Peso Corporal/efeitos dos fármacos , Caquexia/sangue , Caquexia/tratamento farmacológico , Citocinas/sangue , Modelos Animais de Doenças , Quimioterapia Combinada , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazolidinas/administração & dosagem , Mediadores da Inflamação/sangue , Insulina/sangue , Masculino , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculos/patologia , Atrofia Muscular/tratamento farmacológico , Neoplasias/patologia , Tamanho do Órgão , Rosiglitazona , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Tiazolidinedionas/administração & dosagem , Proteínas com Motivo Tripartido , Carga Tumoral , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
16.
J Recept Signal Transduct Res ; 34(1): 21-5, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24094140

RESUMO

We investigated the renoprotective effects of imidapril hydrochloride ((-)-(4 S)-3-[(2 S)-2-[[(1 S)-1-ethoxycarbonyl-3-phenylpropyl] amino] propionyl]-1-methyl-2-oxoimidazolidine-4-carboxylic acid hydrochloride, imidapril), an angiotensin-converting enzyme inhibitor, in a diabetic animal model. We used BKS.Cg-+Lepr(db)/+Lepr(db) (db/db) mice, a genetic animal model of obese type 2 diabetes. Diabetic db/db mice suffered from glomerular hyperfiltration, albuminuria and hypoalbuminemia. Oral administration of 5 mg/kg/day of imidapril for 3 weeks suppressed renal hyperfiltration, reduced albuminuria and normalized hypoalbuminemia. Imidapril did not influence body weights, blood pressure or blood glucose concentrations in db/db mice. Urinary excretion of heparan sulfate (HS) in non-treated 11-week-old db/db mice was significantly lower than that in age-matched non-diabetic db/+m mice. HS is a component of HS proteoglycans, which are present in glomerular basement membranes and glycocalyx of cell surfaces. Reduced urinary HS excretion indicated glomerular HS loss in db/db mice. Imidapril increased urinary excretion of HS to concentrations observed in db/+m mice, indicating that imidapril prevented the loss of renal HS. These results suggest that imidapril ameliorates renal hyperfiltration and loss of renal contents of HS. Improvement of filtration function and maintenance of HS, which is an important structural component of glomeruli, may contribute to renoprotective effects of imidapril.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Heparitina Sulfato/administração & dosagem , Imidazolidinas/administração & dosagem , Obesidade/tratamento farmacológico , Administração Oral , Albuminúria/complicações , Albuminúria/tratamento farmacológico , Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Animais , Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/complicações , Humanos , Hipoalbuminemia/complicações , Hipoalbuminemia/tratamento farmacológico , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/patologia , Camundongos , Camundongos Endogâmicos NOD , Obesidade/complicações , Obesidade/patologia
17.
Expert Opin Pharmacother ; 14(18): 2463-73, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24195786

RESUMO

OBJECTIVE: Aim of this study was to compare the antiproteinuric effect of imidapril (I) and ramipril (R) in diabetic hypertensive patients with microalbuminuria. RESEARCH DESIGN AND METHODS: One hundred and seventy-six patients were randomised to I 10 - 20 mg once daily (od) (n = 88) or R 5 - 10 mg od (n = 88) for 24 weeks. Clinic, ambulatory, central blood pressure (BP), urinary albumin excretion (UAE), plasma Angiotensin II (Ang II), bradykinin and brain natriuretic peptide (BNP) were assessed at baseline and after 6, 12 and 24 weeks. RESULTS: Both I and R produced a similar decrease in clinic, ambulatory and central BP (p < 0.001 vs baseline). Both treatments significantly reduced UAE throughout the study, but the decrease in UAE associated with I was more pronounced, being evident at week 6 (p = 0.05) and maximal at week 24 end-point (-42 vs -29%, p < 0.01). BNP and Ang II levels were similarly reduced by I and R, while bradykinin increased more with R (+132 vs +86%, p < 0.05). CONCLUSIONS: These findings showed that in diabetic hypertensive patients with microalbuminuria, despite equivalent BP-lowering effect, I produced a greater antiproteinuric effect than R, which might be due to different intrinsic molecular properties of the two drugs.


Assuntos
Albuminúria/tratamento farmacológico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipertensão/tratamento farmacológico , Imidazolidinas/uso terapêutico , Ramipril/uso terapêutico , Adulto , Idoso , Albuminas/análise , Albuminúria/complicações , Albuminúria/urina , Angiotensina II/sangue , Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos , Bradicinina/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/urina , Feminino , Humanos , Hipertensão/complicações , Hipertensão/urina , Imidazolidinas/administração & dosagem , Masculino , Pessoa de Meia-Idade , Peptídeo Natriurético Encefálico/sangue , Estudos Prospectivos , Ramipril/administração & dosagem , Resultado do Tratamento
18.
Inflamm Allergy Drug Targets ; 12(3): 178-86, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23360251

RESUMO

BACKGROUND: Allergic rhinitis, one of the most common atopic diseases, is known to be elicited by Th2 cytokine-mediated inflammatory response. We have shown earlier that a polyol pathway enzyme aldose reductase (AR) regulates airway inflammation; however its role in allergic rhinitis is not known. We have investigated the role of AR in mediating pathological symptoms associated with allergic rhinitis in mice. METHODS: The wild-type (WT) mice treated without or with AR inhibitor and AR knock out (AR(-/-)) mice were sensitized by two intraperitoneal injections of ragweed pollen extract (RWE) with adjuvant alum on days 0 and 4 followed by challenge on day 11 and/or 18 and 25. The allergic rhinitis symptoms were assessed by monitoring the nasal scratch, mast cell degranulation and release of tryptase in nasal lavage, infiltration of inflammatory cells, production of inflammatory cytokines and nasal epithelium remodeling. RESULTS: Sensitization and challenge of mice with RWE produced robust and reproducible pathological symptoms of allergic rhinitis as compared to control mice. AR inhibitor, fidarestat administered mice showed markedly reduced early phase response to allergen exposure such as nasal scratches, mast cells degranulation and release of tryptase in the nasal passage as well as late phase response such as inflammatory cell infiltration and release of Th2 type cytokines and nasal epithelial remodeling. Further, prevention of these events in AR(-/-)) mice suggests the role of AR in the mediation of allergic rhinitis. CONCLUSION: These results indicate an important role of AR in the mediation of RWE-induced allergic rhinitis in mice and prevention by AR inhibitor, fidarestat offers a novel therapeutic approach to ameliorate allergic rhinitis.


Assuntos
Aldeído Redutase/metabolismo , Mastócitos/efeitos dos fármacos , Mucosa Nasal/efeitos dos fármacos , Rinite Alérgica Sazonal/prevenção & controle , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Alérgenos/imunologia , Ambrosia , Animais , Degranulação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Humanos , Imidazolidinas/administração & dosagem , Imidazolidinas/farmacologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Nasal/patologia , Pólen/imunologia , Rinite Alérgica Sazonal/enzimologia , Triptases/metabolismo
19.
AAPS J ; 14(4): 657-63, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22711220

RESUMO

Pre-clinical behavioral pharmacology studies supporting indications like analgesia typically consist of at least three different studies; dose-finding, duration of effect, and tolerance-development studies. Pharmacokinetic (PK) plasma samples are generally taken from a parallel group of animals to avoid disruption of the behavioral pharmacodynamic (PD) endpoint. Our objective was to investigate if pre-clinical behavioral pharmacology studies in rats could be performed effectively by combining three studies into a single experimental design and using sparse PK sampling in the same animals as for PD. A refined dosing strategy was applied for a muscarinic agonist, AZD6088, using the rat spinal nerve ligation heat hyperalgesia model. PD measurements were performed on day 1, 3, 5 and 8. Two PK samples per day were taken day 2 and 4. In a separate control group, PD measurements were performed on rats without PK sampling. Data was analyzed using a population approach in NONMEM. The animals produced a consistent and reproducible response irrespective of day of testing suggesting that blood sampling on alternate days did not interfere with the PD responses. A direct concentration-effect relationship with good precision was established and no tolerance development was observed. The new design combining three studies into one and eliminating a satellite PK group realized substantial savings compared to the old design; animal use was reduced by 58% and time required to generate results was reduced by 55%. The design described here delivers substantial savings in animal lives, time, and money whilst still delivering a good quality and precise description of the PKPD relationship.


Assuntos
Determinação de Ponto Final/métodos , Hiperalgesia/tratamento farmacológico , Imidazolidinas/farmacocinética , Modelos Biológicos , Agonistas Muscarínicos/farmacocinética , Piperidinas/farmacocinética , Animais , Redução de Custos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/economia , Avaliação Pré-Clínica de Medicamentos/métodos , Tolerância a Medicamentos , Imidazolidinas/administração & dosagem , Imidazolidinas/farmacologia , Masculino , Agonistas Muscarínicos/administração & dosagem , Agonistas Muscarínicos/farmacologia , Dinâmica não Linear , Piperidinas/administração & dosagem , Piperidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...