Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 886: 173391, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-32745605

RESUMO

Urotensin II (U-II) has been found to be one of the most potent vasoconstrictor (Ames et al., 1999; Bohm et al., 2002) reported till date. U-II exerts its response via activation of a G-protein coupled receptor, Urotensin II receptor(UT). Binding of U-II to UT leads to an instant increase in the inositol phosphate turnover and intracellular Ca2+. Such an instant Ca2+ release and potent vasoconstriction exerted by U-II is expected to have an important role in the progression of cardiac diseases. We have previously shown that UT antagonist DS37001789 prevents U-II induced blood pressure elevation in mice (Nishi et al., 2019) in a dose dependent manner, with potent efficacy at 30 and 100 mg/kg. Further to this, we have also shown that DS37001789 ameliorates mortality in pressure-overload mice with heart failure (Nishi et al., 2020). We therefore conducted an extensive structure-activity relationship studies to identify molecules with superior efficacy. In the present manuscript, we report the identification of two potent, non-peptide small molecule antagonists of Urotensin II receptor (UT), RCI-0879 and RCI-0298 which blocked the action of U-II, both in vitro and in vivo. These molecules were found to be very potent in in vitro Ca2+ and radioligand binding assays using human and mouse UT over-expressing CHO cells. RCI-0879 and RCI-0298 also exhibited superior efficacy in in vivo mouse pressor response model using C57BL/6 mice, compared to our initial molecules (Nishi et al., 2019) and demonstrated ED50 values of 3.2 mg/kg and 6.8 mg/kg respectively. Our findings reported herewith, further strengthen our concept and belief in UT antagonization as a potential therapeutic approach for the management of chronic heart failure.


Assuntos
Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Urotensinas/antagonistas & inibidores , Animais , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Hipertensão/induzido quimicamente , Hipertensão/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
2.
Heliyon ; 6(2): e03352, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32055741

RESUMO

This study was designed to evaluate the effects of DS37001789, a novel and highly potent urotensin II (U-II) receptor (GPR14) antagonist, against mortality, hypertrophy, and cardiac dysfunction in pressure-overload hypertrophy by transverse aortic constriction (TAC) in mice. In addition, we analyzed the phenotype of GPR14 knockout (KO) mice after TAC induction to confirm the contribution of the U-II/GPR14 system. The oral administration of 0.2% DS37001789 to TAC mice for 12 weeks significantly ameliorated the mortality rate and 0.2% DS37001789 for 4 weeks significantly improved cardiac function by pressure-volume analysis. GPR14 expression was significantly upregulated in the left ventricle in the TAC mice treated with 0.2% DS37001789. Moreover, we confirmed that the significant amelioration of mortality was accomplished by the inhibition of cardiac enlargement and the improvement of cardiac function in GPR14 KO mice after TAC surgery. These results suggest that the U-II/GPR14 system contributes to the progression of heart failure and its blockade ameliorates the mortality via improved cardiac function. The U-II/GPR14 system may thus be an attractive target for treating heart failure with pathological cardiac hypertrophy and DS37001789 may be a novel therapeutic agent for heart failure in patients with pressure-overload conditions such as hypertension and aortic valve stenosis.

3.
Heliyon ; 5(3): e01292, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30923760

RESUMO

Fat storage-inducing transmembrane proteins 1 and 2 (FITM1 and FITM2, respectively) are transmembrane endoplasmic/sarcoplasmic reticulum proteins involved in lipid droplet formation. The physiological functions of FITM1 have only been reported in skeletal muscle, while those of FITM2 were analyzed using genetically engineered mice. However, their roles in the heart have not been characterized. To examine their cardiac functions, we analyzed Fitm1- or Fitm2-knockout mice. Neither constitutive Fitm1 (-/-) aged nor heart failure model mice showed significant differences in heart size or function. Fitm2 (-/-) mice exhibited embryonic death, and aged Fitm2 (+/-) mice had shortened left ventricular end-diastolic dimension, and shortened left ventricular end-systolic dimension. However, body weight and ejection fraction of Fitm2 (+/-) mice were similar to those of wild-type littermates. In the chronic heart failure models, Fitm2 (+/-) mice showed significant suppression of increased left ventricular end-diastolic dimension and reduced ejection fraction. These results suggest the involvement of Fitm2 in chronic heart failure, whereas Fitm1 have a minor effect in this context in mice.

4.
J Cardiovasc Pharmacol ; 73(1): 15-21, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30608334

RESUMO

This study was designed to characterize the pharmacological profile of DS37001789, which is a structurally novel piperazine derivative that acts as urotensin II (U-II) receptor antagonist. DS37001789 inhibited [I]-U-II binding to human GPR14, U-II receptor, with an IC50 value 0.9 nM. Its potency was superior to that of ACT-058362, a nonpeptide U-II receptor antagonist whose IC50 was 120 nM. Human U-II-induced vascular contraction was blocked by DS37001789. The dose-response curve of DS37001789 in rats and monkeys did not show species differences, and it shifted to the right without any effects on the maximum vascular response. Moreover, orally administered DS37001789 dose-dependently prevented human U-II-induced blood pressure elevation in mice, and this effect was significant at dose and higher dose (30 and 100 mg/kg), and its potency was superior to that of ACT-058362 (100 mg/kg). These results suggest that DS37001789 is a highly potent U-II receptor antagonist both in vitro and in vivo, with no marked species difference. DS37001789 would be a useful tool to clarify the physiological roles of U-II/GPR14 system. In addition, it can serve as a novel therapeutic agent for diseases in which the U-II/GPR14 system is upregulated, such as hypertension, heart failure, renal dysfunction, and diabetes.


Assuntos
Anti-Hipertensivos/farmacologia , Aorta Torácica/efeitos dos fármacos , Pressão Arterial/efeitos dos fármacos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Urotensinas/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Anti-Hipertensivos/farmacocinética , Aorta Torácica/metabolismo , Células CHO , Cricetulus , Relação Dose-Resposta a Droga , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Bioconjug Chem ; 29(8): 2829-2837, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30107743

RESUMO

Atrial natriuretic peptide (ANP) exerts beneficial pharmacological effects in the treatment of various cardiovascular disorders, such as acute congestive heart failure (ADHF). However, the clinical use of ANP is limited to the continuous intravenous infusion owing to its short half-life (2.4 ± 0.7 min). In the present study, we conjugated the glyco-modified ANP with a monoclonal antibody (mAb) or an Fc via chemo-enzymatic glyco-engineering using EndoS D233Q/Q303L. The most potent derivative SG-ANP-Fc conjugate extended the half-life to 14.9 d and the duration of blood pressure lowering effect to over 28 d. This new biologic modality provides an opportunity to develop outpatient therapy after ADHF.


Assuntos
Fator Natriurético Atrial/farmacologia , Fator Natriurético Atrial/farmacocinética , Animais , Anticorpos Monoclonais/química , Fator Natriurético Atrial/síntese química , Fator Natriurético Atrial/química , Células CHO , Cricetulus , GMP Cíclico/agonistas , GMP Cíclico/sangue , Glicosilação , Meia-Vida , Humanos , Imunoconjugados/sangue , Macaca fascicularis , Masculino , Ratos , Ratos Wistar
6.
J Pharmacol Exp Ther ; 363(3): 402-410, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29021382

RESUMO

Heart failure often presents with prognosis-relevant impaired renal function. To investigate whether the chronic activation of guanylate cyclase-A (GC-A) protects both heart and kidney, we examined the effects of TDT, a neprilysin (NEP)-resistant natriuretic peptide (NP) derivative, on cardiac and renal dysfunction in Dahl salt-sensitive hypertensive (DS) rats. Pretreatment with NEP or NEP inhibitor did not influence GC-A activation by TDT both in vitro and in vivo, resulting in a long-acting profile of TDT compared with native human atrial NP (hANP). The repeated administration of TDT to DS rats suppressed the progress of cardiac hypertrophy, systolic/diastolic dysfunction, and proteinuria in a dose-dependent manner. Compared with vehicle and hANP, salt diet-induced podocyte injury was reduced by TDT, as analyzed by urinary podocalyxin concentration, renal expression of nephrin mRNA, and glomerular expression of desmin protein. Since glomerular TRPC6 plays detrimental roles in podocyte homeostasis, we examined the renal expression of TRPC6 in DS rats and found that salt diet upregulated the expression of TRPC6. Importantly, TRPC6 induction was significantly decreased in TDT-treated rats, compared with vehicle and hANP. Consistently, in primary-culture podocytes from DS rats, TDT inhibited ATP-induced calcium influx, similar to TRPC inhibitor SKF96365. Finally, TDT-mediated protection of podocytes was abolished by protein kinase G inhibitor KT5823. In conclusion, TDT treatment attenuated heart and kidney dysfunction, accompanied by podocyte protection through inhibition of TRPC6. Thus, long-acting NPs could be a new avenue for treatment of heart failure.


Assuntos
Fator Natriurético Atrial/uso terapêutico , Coração/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Rim/efeitos dos fármacos , Receptores do Fator Natriurético Atrial/metabolismo , Sequência de Aminoácidos , Animais , Fator Natriurético Atrial/genética , Células CHO , Técnicas de Cultura de Células , Cricetulus , GMP Cíclico/sangue , Relação Dose-Resposta a Droga , Coração/fisiopatologia , Humanos , Hipertensão/enzimologia , Hipertensão/fisiopatologia , Rim/fisiopatologia , Podócitos/efeitos dos fármacos , Podócitos/patologia , Ratos Endogâmicos Dahl , Receptores do Fator Natriurético Atrial/genética , Proteínas Recombinantes
7.
Bioorg Med Chem Lett ; 27(21): 4904-4907, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28958620

RESUMO

Novel thienopyrimidine compounds 2 and 3 were discovered from high-throughput screening as Natriuretic Peptide Receptor A (NPR-A) agonists. Scaffold hopping of a thienopyrimidine ring to a quinazoline ring, introduction of the basic functional group and optimization of the substituent on the 6-position of the benzene ring of quinazoline led to improved agonistic activity. We discovered compound 48, which showed potent agonistic activity for NPR-A with an EC50 value of 0.073µM, indicating 350-fold potency compared to the hit compound 3.


Assuntos
Pirimidinas/metabolismo , Receptores do Fator Natriurético Atrial/agonistas , Animais , Células CHO , Cricetinae , Cricetulus , Avaliação Pré-Clínica de Medicamentos , Humanos , Pirimidinas/síntese química , Pirimidinas/química , Quinazolinas/síntese química , Quinazolinas/química , Quinazolinas/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Relação Estrutura-Atividade
8.
Cell Death Discov ; 3: 17045, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28725491

RESUMO

In cardiac myocytes, regulation of mitochondrial Ca2+ is important for cellular signaling and cardiac contraction. Ca2+ entry into the mitochondria is mediated by a highly selective Ca2+ channel called the mitochondrial calcium uniporter, which consists of a pore-forming subunit MCU and regulatory subunits such as MICU1. Although pharmacological regulation of the mitochondrial Ca2+ influx is a promising approach to controlling the cellular functions, a cell-permeable and specific inhibitor of the mitochondrial calcium uniporter has not yet been developed. Here, we identify a novel cell-permeable inhibitor of the uniporter by a high-throughput screening of 120 000 small-molecule compounds. In our study, DS16570511 dose-dependently inhibited serum-induced mitochondrial Ca2+ influx in HEK293A cells with an IC50 of 7 µM. DS16570511 inhibited Ca2+ uptake of isolated mitochondria from human cells, rat heart and pig heart. Overexpression of hMCU or hMICU1 in HEK293A cells increased mitochondrial Ca2+ influx, and the increases were completely suppressed by the pretreatment with DS16570511. DS16570511 also blocks mitochondrial Ca2+ overload in a Langendorff perfused beating rat heart. Interestingly, DS16570511 increased cardiac contractility without affecting heart rate in the perfused heart. These results show that DS16570511 is a novel cell-permeable inhibitor of the mitochondrial calcium uniporter and applicable for control of the cardiac functions.

9.
Eur J Pharmacol ; 795: 58-65, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27915044

RESUMO

It has been elucidated that mineralocorticoid receptor antagonists reduce mortality in patients with congestive heart failure and post-acute myocardial infarction. A direct inhibition of aldosterone synthase (CYP11B2) is also expected to have therapeutic benefits equal in quality to mineralocorticoid receptor antagonists in terms of reducing mineralocorticoid receptor signaling. Therefore, we have screened our chemical libraries and identified a novel and potent aldosterone synthase inhibitor, 2,2,2-trifluoro-1-{4-[(4-fluorophenyl)amino]pyrimidin-5-y}-1-[1-(methylsulfonyl)piperidin-4-yl]ethanol (compound 1), by lead optimization. Pharmacological properties of compound 1 were examined in in vitro cell-based assays and an in vivo mouse model of pressure-overload hypertrophy by transverse aortic constriction (TAC). Compound 1 showed potent CYP11B2 inhibition against human and mouse enzymes (IC50; 0.003µM and 0.096µM, respectively) in a cell-based assay. The oral administration of 0.06% compound 1 in the food mixture of a mouse TAC model significantly reduced the plasma aldosterone level and ameliorated mortality rate. This study is the first to demonstrate that a CYP11B2 inhibitor improved survival rates of heart failure induced by pressure-overload in mice. The treatment of 0.06% compound 1 did not elevate plasma potassium level in this model, although further evaluation of hyperkalemia is needed. These results suggest that compound 1 can be developed as a promising oral CYP11B2 inhibitor for pharmaceutical applications. Compound 1 could also be a useful compound for clarifying the role of aldosterone in cardiac hypertrophy.


Assuntos
Citocromo P-450 CYP11B2/antagonistas & inibidores , Inibidores das Enzimas do Citocromo P-450/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/mortalidade , Pressão/efeitos adversos , Aldosterona/biossíntese , Animais , Cardiomegalia/complicações , Cardiomegalia/tratamento farmacológico , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Inibidores das Enzimas do Citocromo P-450/uso terapêutico , Células HEK293 , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Humanos , Camundongos , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico
10.
J Clin Invest ; 124(6): 2464-71, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24837433

RESUMO

Inhibition of cGMP-specific phosphodiesterase 5 (PDE5) ameliorates pathological cardiac remodeling and has been gaining attention as a potential therapy for heart failure. Despite promising results in males, the efficacy of the PDE5 inhibitor sildenafil in female cardiac pathologies has not been determined and might be affected by estrogen levels, given the hormone's involvement in cGMP synthesis. Here, we determined that the heart-protective effect of sildenafil in female mice depends on the presence of estrogen via a mechanism that involves myocyte eNOS-dependent cGMP synthesis and the cGMP-dependent protein kinase Iα (PKGIα). Sildenafil treatment failed to exert antiremodeling properties in female pathological hearts from Gαq-overexpressing or pressure-overloaded mice after ovary removal; however, estrogen replacement restored the effectiveness of sildenafil in these animals. In females, sildenafil-elicited myocardial PKG activity required estrogen, which stimulated tonic cardiomyocyte cGMP synthesis via an eNOS/soluble guanylate cyclase pathway. In contrast, eNOS activation, cGMP synthesis, and sildenafil efficacy were not estrogen dependent in male hearts. Estrogen and sildenafil had no impact on pressure-overloaded hearts from animals expressing dysfunctional PKGIα, indicating that PKGIα mediates antiremodeling effects. These results support the importance of sex differences in the use of PDE5 inhibitors for treating heart disease and the critical role of estrogen status when these agents are used in females.


Assuntos
Estrogênios/metabolismo , Cardiopatias/tratamento farmacológico , Cardiopatias/metabolismo , Inibidores da Fosfodiesterase 5/farmacologia , Animais , Cardiotônicos/farmacologia , GMP Cíclico/biossíntese , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Modelos Animais de Doenças , Estradiol/administração & dosagem , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Guanilato Ciclase/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Ovariectomia , Piperazinas/farmacologia , Purinas/farmacologia , Receptores do Fator Natriurético Atrial/deficiência , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo , Caracteres Sexuais , Citrato de Sildenafila , Sulfonas/farmacologia , Resultado do Tratamento
11.
Antioxid Redox Signal ; 20(2): 267-80, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23581564

RESUMO

AIMS: Monoamine oxidases (MAOs) are mitochondrial flavoenzymes responsible for neurotransmitter and biogenic amines catabolism. MAO-A contributes to heart failure progression via enhanced norepinephrine catabolism and oxidative stress. The potential pathogenetic role of the isoenzyme MAO-B in cardiac diseases is currently unknown. Moreover, it is has not been determined yet whether MAO activation can directly affect mitochondrial function. RESULTS: In wild type mice, pressure overload induced by transverse aortic constriction (TAC) resulted in enhanced dopamine catabolism, left ventricular (LV) remodeling, and dysfunction. Conversely, mice lacking MAO-B (MAO-B(-/-)) subjected to TAC maintained concentric hypertrophy accompanied by extracellular signal regulated kinase (ERK)1/2 activation, and preserved LV function, both at early (3 weeks) and late stages (9 weeks). Enhanced MAO activation triggered oxidative stress, and dropped mitochondrial membrane potential in the presence of ATP synthase inhibitor oligomycin both in neonatal and adult cardiomyocytes. The MAO-B inhibitor pargyline completely offset this change, suggesting that MAO activation induces a latent mitochondrial dysfunction, causing these organelles to hydrolyze ATP. Moreover, MAO-dependent aldehyde formation due to inhibition of aldehyde dehydrogenase 2 activity also contributed to alter mitochondrial bioenergetics. INNOVATION: Our study unravels a novel role for MAO-B in the pathogenesis of heart failure, showing that both MAO-driven reactive oxygen species production and impaired aldehyde metabolism affect mitochondrial function. CONCLUSION: Under conditions of chronic hemodynamic stress, enhanced MAO-B activity is a major determinant of cardiac structural and functional disarrangement. Both increased oxidative stress and the accumulation of aldehyde intermediates are likely liable for these adverse morphological and mechanical changes by directly targeting mitochondria.


Assuntos
Mitocôndrias Cardíacas/metabolismo , Monoaminoxidase/metabolismo , Miócitos Cardíacos/metabolismo , Disfunção Ventricular/metabolismo , Disfunção Ventricular/fisiopatologia , Aldeídos/metabolismo , Animais , Apoptose/genética , Pressão Sanguínea , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Dopamina/metabolismo , Ativação Enzimática , Fibrose , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monoaminoxidase/genética , Miócitos Cardíacos/patologia , Oxirredução , Estresse Oxidativo/genética , Fosforilação , Ratos , Espécies Reativas de Oxigênio/metabolismo , Disfunção Ventricular/genética , Função Ventricular Esquerda
12.
Bioorg Med Chem ; 21(18): 5907-22, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23886807

RESUMO

We report synthesis and optimization of a series of (3S,5R)-5-(2,2-dimethyl-5-oxo-4-phenylpiperazin-1-yl)piperidine-3-carboxamides as renin inhibitors. Chemical modification of P1', P2' and P3 portions led to a promising 3,5-disubstituted piperidine 32o showing high renin inhibitory activity and favorable oral exposure in both rats and cynomolgus monkeys with acceptable CYP and hERG current inhibition. Compound 32o exhibited a significant blood pressure lowering effect by oral administration in two hypertensive animal models, double transgenic rats and furosemide pretreated cynomolgus monkeys.


Assuntos
Amidas/química , Piperazinas/síntese química , Piperidinas/química , Piperidinas/síntese química , Inibidores de Proteases/síntese química , Renina/antagonistas & inibidores , Administração Oral , Amidas/farmacocinética , Amidas/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Furosemida/farmacologia , Meia-Vida , Hipertensão/tratamento farmacológico , Macaca fascicularis , Piperazinas/farmacocinética , Piperazinas/uso terapêutico , Piperidinas/farmacocinética , Piperidinas/uso terapêutico , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/uso terapêutico , Ratos , Ratos Transgênicos , Renina/metabolismo , Relação Estrutura-Atividade
13.
Bioorg Med Chem ; 21(11): 3175-96, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23598247

RESUMO

With the aim to address an undesired cardiac issue observed with our related compound in the recently disclosed novel series of renin inhibitors, further chemical modifications of this series were performed. Extensive structure-activity relationships studies as well as in vivo cardiac studies using the electrophysiology rat model led to the discovery of clinical candidate trans-adamantan-1-ol analogue 56 (DS-8108b) as a potent renin inhibitor with reduced potential cardiac risk. Oral administration of single doses of 3 and 10 mg/kg of 56 in cynomolgus monkeys pre-treated with furosemide led to significant reduction of mean arterial blood pressure for more than 12 h.


Assuntos
Anti-Hipertensivos/síntese química , Arritmias Cardíacas/prevenção & controle , Coração/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Piperazinas/síntese química , Inibidores de Proteases/síntese química , Renina/antagonistas & inibidores , Administração Oral , Animais , Anti-Hipertensivos/farmacocinética , Anti-Hipertensivos/farmacologia , Pressão Arterial/efeitos dos fármacos , Feminino , Coração/fisiopatologia , Humanos , Hipertensão/enzimologia , Hipertensão/fisiopatologia , Macaca fascicularis , Masculino , Técnicas de Cultura de Órgãos , Piperazinas/farmacocinética , Piperazinas/farmacologia , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/farmacologia , Coelhos , Ratos , Renina/química , Renina/metabolismo , Relação Estrutura-Atividade
15.
Circulation ; 126(8): 942-51, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22829024

RESUMO

BACKGROUND: In the normal heart, phosphodiesterase type 5 (PDE5) hydrolyzes cGMP coupled to nitric oxide- (specifically from nitric oxide synthase 3) but not natriuretic peptide (NP)-stimulated guanylyl cyclase. PDE5 is upregulated in hypertrophied and failing hearts and is thought to contribute to their pathophysiology. Because nitric oxide signaling declines whereas NP-derived cGMP rises in such diseases, we hypothesized that PDE5 substrate selectivity is retargeted to blunt NP-derived signaling. METHODS AND RESULTS: Mice with cardiac myocyte inducible PDE5 overexpression (P5(+)) were crossed to those lacking nitric oxide synthase 3 (N3(-)), and each model, the double cross, and controls were subjected to transaortic constriction. P5(+) mice developed worse dysfunction and hypertrophy and enhanced NP stimulation, whereas N3(-) mice were protected. However, P5(+)/N3(-) mice behaved similarly to P5(+) mice despite the lack of nitric oxide synthase 3-coupled cGMP generation, with protein kinase G activity suppressed in both models. PDE5 inhibition did not alter atrial natriuretic peptide-stimulated cGMP in the resting heart but augmented it in the transaortic constriction heart. This functional retargeting was associated with PDE5 translocation from sarcomeres to a dispersed distribution. P5(+) hearts exhibited higher oxidative stress, whereas P5(+)/N3(-) hearts had low levels (likely owing to the absence of nitric oxide synthase 3 uncoupling). This highlights the importance of myocyte protein kinase G activity as a protection for pathological remodeling. CONCLUSIONS: These data provide the first evidence for functional retargeting of PDE5 from one compartment to another, revealing a role for natriuretic peptide-derived cGMP hydrolysis by this esterase in diseased heart myocardium. Retargeting likely affects the pathophysiological consequence and the therapeutic impact of PDE5 modulation in heart disease.


Assuntos
Fator Natriurético Atrial/metabolismo , Cardiomegalia/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais/fisiologia , Animais , Cardiomegalia/patologia , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Modelos Animais de Doenças , Feminino , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/enzimologia , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Óxido Nítrico Sintase Tipo III/genética , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Remodelação Ventricular/fisiologia
16.
Bioorg Med Chem Lett ; 22(14): 4561-6, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22726934

RESUMO

Introduction of the 2,2-dimethyl-4-phenylpiperazin-5-one scaffold into the P(3)-P(1) portion of the (2S,4S,5S)-5-amino-6-dialkylamino-4-hydroxy-2-isopropylhexanamide backbone dramatically increased the renin inhibitory activity without using the interaction to the S(3)(sp) pocket. Compound 31 exhibited >10,000-fold selectivity over other human proteases, and 18.5% oral bioavailability in monkey.


Assuntos
Amidas/química , Piperazinas/química , Renina/antagonistas & inibidores , Amidas/farmacologia , Aminação , Desenho de Fármacos , Hidroxilação , Metilação , Modelos Moleculares , Piperazina , Relação Estrutura-Atividade
17.
ACS Med Chem Lett ; 3(9): 754-8, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900544

RESUMO

A novel orally bioavailable renin inhibitor, DS-8108b (5), showing potent renin inhibitory activity and excellent in vivo efficacy is described. We report herein the synthesis and pharmacological effects of 5 including renin inhibitory activity in vitro, suppressive effects of ex vivo plasma renin activity (PRA) in cynomolgus monkey, pharmacokinetic data, and blood pressure-lowering effects in an animal model. Compound 5 demonstrated inhibitory activities toward human renin (IC50 = 0.9 nM) and human and monkey PRA (IC50 = 1.9 and 6.3 nM, respectively). Oral administration of single doses of 3 and 10 mg/kg of 5 in cynomolgus monkey on pretreatment with furosemide led to dose-dependent significant reductions in ex vivo PRA and sustained lowering of mean arterial blood pressure for more than 12 h.

18.
Nature ; 477(7363): 188-90, 2011 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-21866100

RESUMO

The nuclear bulge is a region with a radius of about 200 parsecs around the centre of the Milky Way. It contains stars with ages ranging from a few million years to over a billion years, yet its star-formation history and the triggering process for star formation remain to be resolved. Recently, episodic star formation, powered by changes in the gas content, has been suggested. Classical Cepheid variable stars have pulsation periods that decrease with increasing age, so it is possible to probe the star-formation history on the basis of the distribution of their periods. Here we report the presence of three classical Cepheids in the nuclear bulge with pulsation periods of approximately 20 days, within 40 parsecs (projected distance) of the central black hole. No Cepheids with longer or shorter periods were found. We infer that there was a period about 25 million years ago, and possibly lasting until recently, in which star formation increased relative to the period of 30-70 million years ago.

19.
J Am Coll Cardiol ; 56(24): 2021-30, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-20970280

RESUMO

OBJECTIVES: we tested the hypothesis that bi-directional, gene-targeted regulation of cardiomyocyte cyclic guanosine monophosphate-selective phosphodiesterase type 5 (PDE5) influences maladaptive remodeling in hearts subjected to sustained pressure overload. BACKGROUND: PDE5 expression is up-regulated in human hypertrophied and failing hearts, and its inhibition (e.g., by sildenafil) stimulates protein kinase G activity, suppressing and reversing maladaptive hypertrophy, fibrosis, and contractile dysfunction. Sildenafil is currently being clinically tested for the treatment of heart failure. However, researchers of new studies have questioned the role of myocyte PDE5 and protein kinase G (PKG) to this process, proposing alternative targets and mechanisms. METHODS: mice with doxycycline-controllable myocyte-specific PDE5 gene expression were generated (medium transgenic [TG] and high TG expression lines) and subjected to sustained pressure overload. RESULTS: Rest myocyte and heart function, histology, and molecular profiling were normal in both TG lines versus controls at 2 months of age. However, upon exposure to pressure overload (aortic banding), TG hearts developed more eccentric remodeling, maladaptive molecular signaling, depressed function, and amplified fibrosis with up-regulation of tissue growth factor signaling pathways. PKG activation was inhibited in TG myocytes versus controls. After establishing a severe cardiomyopathic state, high-TG mice received doxycycline to suppress PDE5 expression/activity only in myocytes. This in turn enhanced PKG activity and reversed all previously amplified maladaptive responses, despite sustained pressure overload. Sildenafil was also effective in this regard. CONCLUSIONS: these data strongly support a primary role of myocyte PDE5 regulation to myocardial pathobiology and PDE5 targeting therapy in vivo and reveal a novel mechanism of myocyte-orchestrated extracellular matrix remodeling via PDE5/cyclic guanosine monophosphate-PKG regulatory pathways.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Regulação Enzimológica da Expressão Gênica , Monócitos/metabolismo , Miócitos Cardíacos/metabolismo , Remodelação Ventricular/genética , Animais , Humanos , Camundongos , Transdução de Sinais
20.
Biol Pharm Bull ; 33(9): 1500-5, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20823564

RESUMO

A sphingosine-1-phosphate receptor 1 (S1P1) antagonist is expected to be an anti-angiogenic compound; however, there are few reports that demonstrated that a S1P1 inhibitor improved the disease state in an angiogenic animal model. Since we determined that a prototype S1P1 antagonist was an in vivo angiogenesis inhibitor, we developed the derivatives to acquire more effective compounds. In this report, we show the S1P1 antagonistic activity of some representatives, especially compound 5 {sodium 4-[(4-butoxyphenyl)thio]-2'-[{4-[(heptylthio)methyl]-2-hydroxyphenyl}(hydroxy)methyl]biphenyl-3-sulfonate}. The IC50 values calculated from an intracellular cyclic AMP measurement assay and a [33P]sphingosine-1-phosphate (Sph-1-P)/S1P1 binding assay were 38 and 200 nM, respectively. A subtype specificity test for the other Sph-1-P receptors showed that compound 5 was the S1P1-directional antagonist. It also inhibited the proliferation, migration, and tube formation of human umbilical vein endothelial cells stimulated by Sph-1-P with the IC50 values of 18, 650, and 230 nM, respectively. A cytotoxicity assay concurrently performed with a tube formation assay supported the hypothesis that these biological effects were not due to its cytotoxicity. Furthermore, administration (10 mg/kg, intravenously) to anesthetized Sprague-Dawley rats inhibited Sph-1-P-induced hypotension by 100-90% for 30 min. This is presumably through the inhibition of Sph-1-P-induced vasorelaxation, mainly by the blocking of S1P1 and/or S1P3. Taken together, these results show that compound 5 is an inhibitor of in vitro and in vivo Sph-1-P signaling, and that it will be useful to elucidate the in vivo effect of Sph-1-P on vascular endothelial cells.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Lisofosfolipídeos/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Esfingosina/análogos & derivados , Vasodilatação/efeitos dos fármacos , Animais , Células CHO , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Cricetinae , Cricetulus , Células Endoteliais/fisiologia , Humanos , Lisofosfolipídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/antagonistas & inibidores , Esfingosina/farmacologia , Vasodilatação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...