Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 382(3): 266-276, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35779860

RESUMO

Diabetic nephropathy is a leading cause of end-stage renal disease, characterized by endothelial dysfunction and a compromised glomerular permeability barrier. Dysregulation of the angiopoietin 1 (ANGPT1)/angiopoietin 2 (ANGPT2) signaling axis is implicated in disease progression. We recently described the discovery of an IgG1 antibody, O010, with therapeutic potential to elevate circulating endogenous ANGPT1, a tyrosine kinase with Ig and epidermal growth factor (EGF) homology domains-2 (TIE2) agonist. Studies are described that detail the effect of various ANGPT1-elevating strategies to limit progression of renal dysfunction in diabetic-obese (db/db) mice. Results demonstrate that adeno-associated virus- or DNA minicircle-directed overexpression of ANGPT1 elicits a reduction in albuminuria (56%-73%) and an improvement in histopathology score (18% reduction in glomerulosclerosis). An improved acetylcholine response in isolated aortic rings was also observed indicative of a benefit on vascular function. In separate pharmacokinetic studies, an efficacious dose of the ANGPT1 DNA minicircle increased circulating levels of the protein by >80%, resulting in a concomitant suppression of ANGPT2. At a dose of O010-producing maximal elevation of circulating ANGPT1 achievable with the molecule (60% increase), no suppression of ANGPT2 was observed in db/db mice, suggesting insufficient pathway engagement; no reduction in albuminuria or improvement in histopathological outcomes were observed. To pinpoint the mechanism resulting in lack of efficacy, we demonstrate, using confocal microscopy, an interference with TIE2 translocation to adherens junctions, resulting in a loss of protection against vascular permeability normally conferred by ANGPT1. Results demonstrated the essential importance of ANGPT1 to maintain the glomerular permeability barrier, and, due to interference of O010 with this process, led to the discontinuation of the molecule for clinical development. SIGNIFICANCE STATEMENT: This body of original research demonstrates that elevation of systemic angiopoietin 1 (ANGPT1) is protective against diabetic nephropathy. However, using a novel biotherapeutic approach to elevate systemic ANGPT1 renoprotection was not observed; we demonstrate that protection was lost due to interference of the therapeutic with ANGPT1/ tyrosine kinase with Ig and EGF homology domains-2 translocation to adherens junctions. Thus, the clinical development of the antibody was terminated.


Assuntos
Angiopoietina-1 , Diabetes Mellitus , Nefropatias Diabéticas , Albuminúria , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Fator de Crescimento Epidérmico , Camundongos , Camundongos Obesos , Proteínas Tirosina Quinases
2.
PLoS One ; 9(8): e105883, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170619

RESUMO

Inflammation is associated with immune cells infiltrating into the inflammatory site and pain. CC chemokine receptor 1 (CCR1) mediates trafficking of leukocytes to sites of inflammation. However, the contribution of CCR1 to pain is incompletely understood. Here we report an unexpected discovery that CCR1-mediated trafficking of neutrophils and CCR1 activity on non-hematopoietic cells both modulate pain. Using a genetic approach (CCR1-/- animals) and pharmacological inhibition of CCR1 with selective inhibitors, we show significant reductions in pain responses using the acetic acid-induced writhing and complete Freund's adjuvant-induced mechanical hyperalgesia models. Reductions in writhing correlated with reduced trafficking of myeloid cells into the peritoneal cavity. We show that CCR1 is highly expressed on circulating neutrophils and their depletion decreases acetic acid-induced writhing. However, administration of neutrophils into the peritoneal cavity did not enhance acetic acid-induced writhing in wild-type (WT) or CCR1-/- mice. Additionally, selective knockout of CCR1 in either the hematopoietic or non-hematopoietic compartments also reduced writhing. Together these data suggest that CCR1 functions to significantly modulate pain by controlling neutrophil trafficking to the inflammatory site and having an unexpected role on non-hematopoietic cells. As inflammatory diseases are often accompanied with infiltrating immune cells at the inflammatory site and pain, CCR1 antagonism may provide a dual benefit by restricting leukocyte trafficking and reducing pain.


Assuntos
Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Dor/imunologia , Receptores CCR1/imunologia , Ácido Acético , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Movimento Celular/genética , Movimento Celular/imunologia , Citometria de Fluxo , Adjuvante de Freund , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/imunologia , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Infiltração de Neutrófilos/genética , Neutrófilos/metabolismo , Dor/induzido quimicamente , Dor/genética , Medição da Dor/métodos , Peritonite/genética , Peritonite/imunologia , Peritonite/metabolismo , Receptores CCR1/antagonistas & inibidores , Receptores CCR1/genética
3.
J Pharmacol Exp Ther ; 348(3): 421-31, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24399854

RESUMO

Bile acids (BAs) and BA receptors, including G protein-coupled bile acid receptor 1 (GPBAR1), represent novel targets for the treatment of metabolic and inflammatory disorders. However, BAs elicit myriad effects on cardiovascular function, although this has not been specifically ascribed to GPBAR1. This study was designed to test whether stimulation of GPBAR1 elicits effects on cardiovascular function that are mechanism based that can be identified in acute ex vivo and in vivo cardiovascular models, to delineate whether effects were due to pathways known to be modulated by BAs, and to establish whether a therapeutic window between in vivo cardiovascular liabilities and on-target efficacy could be defined. The results demonstrated that the infusion of three structurally diverse and selective GPBAR1 agonists produced marked reductions in vascular tone and blood pressure in dog, but not in rat, as well as reflex tachycardia and a positive inotropic response, effects that manifested in an enhanced cardiac output. Changes in cardiovascular function were unrelated to modulation of the levothyroxine/thyroxine axis and were nitric oxide independent. A direct effect on vascular tone was confirmed in dog isolated vascular rings, whereby concentration-dependent decreases in tension that were tightly correlated with reductions in vascular tone observed in vivo and were blocked by iberiotoxin. Compound concentrations in which cardiovascular effects occurred, both ex vivo and in vivo, could not be separated from those necessary for modulation of GPBAR1-mediated efficacy, resulting in project termination. These results are the first to clearly demonstrate direct and potent peripheral arterial vasodilation due to GPBAR1 stimulation in vivo through activation of large conductance Ca(2+) activated potassium channel K(Ca)1.1.


Assuntos
Artérias/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Vasodilatação/efeitos dos fármacos , Animais , Artérias/fisiologia , Fator Natriurético Atrial/sangue , Células CHO , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/patologia , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Citocinas/sangue , Dinitrofluorbenzeno/análogos & derivados , Cães , Endotelina-1/sangue , Humanos , Imidazóis/farmacologia , Técnicas In Vitro , Masculino , Óxido Nítrico/biossíntese , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Tiroxina/sangue , Triazóis/farmacologia
4.
J Pharmacol Exp Ther ; 340(3): 492-500, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22128344

RESUMO

We previously reported the discovery of a novel ribosomal S6 kinase 2 (RSK2) inhibitor, (R)-5-Methyl-1-oxo-2,3,4,5-tetrahydro-1H-[1,4]diazepino[1,2-a] indole-8-carboxylic acid [1-(3-dimethylamino-propyl)-1H-benzoimidazol-2-yl]-amide (BIX 02565), with high potency (IC(50) = 1.1 nM) targeted for the treatment of heart failure. In the present study, we report that despite nanomolar potency at the target, BIX 02565 elicits off-target binding at multiple adrenergic receptor subtypes that are important in the control of vascular tone and cardiac function. To elucidate in vivo the functional consequence of receptor binding, we characterized the cardiovascular (CV) profile of the compound in an anesthetized rat CV screen and telemetry-instrumented conscious rats. Infusion of BIX 02565 (1, 3, and 10 mg/kg) in the rat CV screen resulted in a precipitous decrease in both mean arterial pressure (MAP; to -65 ± 6 mm Hg below baseline) and heart rate (-93 ± 13 beats/min). In telemetry-instrumented rats, BIX 02565 (30, 100, and 300 mg/kg p.o. QD for 4 days) elicited concentration-dependent decreases in MAP after each dose (to -39 ± 4 mm Hg on day 4 at T(max)); analysis by Demming regression demonstrated strong correlation independent of route of administration and influence of anesthesia. Because of pronounced off-target effects of BIX 02565 on cardiovascular function, a high-throughput selectivity screen at adrenergic α(1A) and α(2A) was performed for 30 additional RSK2 inhibitors in a novel chemical series; a wide range of adrenergic binding was achieved (0-92% inhibition), allowing for differentiation within the series. Eleven lead compounds with differential binding were advanced to the rat CV screen for in vivo profiling. This led to the identification of potent RSK2 inhibitors (cellular IC(50) <0.14 nM) without relevant α(1A) and α(2A) inhibition and no adverse cardiovascular effects in vivo.


Assuntos
Azepinas/farmacologia , Benzimidazóis/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Descoberta de Drogas , Masculino , Ratos , Ratos Sprague-Dawley
5.
J Pharmacol Exp Ther ; 329(3): 928-37, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19255283

RESUMO

ABT-869 [N-(4-(3-amino-1H-indazol-4-yl)phenyl)-N'-(2-fluoro-5-methylphenyl)urea] is a novel multitargeted inhibitor of the vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinase family members. ABT-869 demonstrates tumor growth inhibition in multiple preclinical animal models and in early clinical trials. VEGF receptor inhibition is also associated with reversible hypertension that may limit its benefit clinically. To evaluate optimal therapeutic approaches to prevent hypertension with VEGF receptor inhibition, we characterized the dose-dependent effects of seven antihypertensive agents from three mechanistic classes [angiotensin-converting enzyme inhibitors (ACEis), angiotensin receptor blockers (ARBs), calcium channel blockers (CCBs)] on hypertension induced by ABT-869 in conscious telemetry rats. We report that ABT-869-induced hypertension can be prevented and reversed with subtherapeutic or therapeutic doses of antihypertensive drugs with a general rank order of ACEi > ARB > CCB. In SCID mice, the ACE inhibitor, enalapril (C(20)H(28)N(2)O(5) x C(4)H(4)O(4)) at 30 mg/kg, prevented hypertension, with no attenuation of the antitumor efficacy of ABT-869. These studies demonstrate that the adverse cardiovascular effects of the VEGF/PDGF receptor tyrosine kinase inhibitor, ABT-869, are readily controlled by conventional antihypertensive therapy without affecting antitumor efficacy.


Assuntos
Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Indazóis/farmacologia , Neoplasias/tratamento farmacológico , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Acrilatos/farmacologia , Anlodipino/farmacologia , Antagonistas de Receptores de Angiotensina , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Enalapril/farmacologia , Humanos , Imidazóis/farmacologia , Indazóis/efeitos adversos , Indazóis/uso terapêutico , Lisinopril/farmacologia , Masculino , Camundongos , Camundongos SCID , Neoplasias/patologia , Nifedipino/farmacologia , Compostos de Fenilureia/efeitos adversos , Compostos de Fenilureia/uso terapêutico , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/uso terapêutico , Ramipril/farmacologia , Ratos , Ratos Sprague-Dawley , Telmisartan , Tiofenos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Cardiovasc Pharmacol ; 53(2): 173-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19188829

RESUMO

ABT-869 is a novel multitargeted inhibitor of vascular endothelial growth factor and platelet-derived growth factor receptor tyrosine kinases (RTKs) with potent antiangiogenic properties that slow tumor progression. Vascular endothelial growth factor receptor blockade has been shown to produce hypertension. Atrasentan is a potent and selective endothelin (ETA) receptor antagonist that lowers blood pressure and affects tumor growth. To assess the utility of ETA receptor blockade in controlling hypertension with RTK inhibition, we evaluated the ability of atrasentan to block hypertension with ABT-869 in conscious, telemetry-instrumented rats. Changes in mean arterial pressure (MAP) and heart rate (HR) were evaluated using mean values and the area under the curve (AUC). Atrasentan (0.5, 1.5, and 5.0 mg kg(-1) d(-1) for 5 days) elicited dose-dependent decreases in MAP-AUC (-16.7 +/- 1.3, -20.94 +/- 3.68, and -30.12 +/- 3.57 mm Hg x day, respectively) compared with vehicle. ABT-869 (1, 3, 10, 30 mg kg(-1) d(-1) for 5 days) increased MAP compared with vehicle (MAP-AUC values of -5.52 +/- 3.75, 12.7 +/- 8.4, 37.5 +/- 4.4, and 63.8 +/- 3.3 mm Hg x day, respectively). Pretreatment with atrasentan (5 mg/kg for 5 days) prevented and abolished the hypertensive effects of ABT-869. Thus, ETA receptor blockade effectively alleviated hypertension with RTK inhibition and may serve a dual therapeutic role by preventing hypertension and slowing tumor progression.


Assuntos
Inibidores da Angiogênese/farmacologia , Antagonistas do Receptor de Endotelina A , Hipertensão/prevenção & controle , Indazóis/farmacologia , Compostos de Fenilureia/farmacologia , Pirrolidinas/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Inibidores da Angiogênese/efeitos adversos , Animais , Área Sob a Curva , Atrasentana , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Indazóis/efeitos adversos , Masculino , Compostos de Fenilureia/efeitos adversos , Pirrolidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Telemetria
7.
Pain ; 142(1-2): 27-35, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19135797

RESUMO

Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel that functions as an integrator of multiple pain stimuli including heat, acid, capsaicin and a variety of putative endogenous lipid ligands. TRPV1 antagonists have been shown to decrease inflammatory pain in animal models and to produce limited hyperthermia at analgesic doses. Here, we report that ABT-102, which is a potent and selective TRPV1 antagonist, is effective in blocking nociception in rodent models of inflammatory, post-operative, osteoarthritic, and bone cancer pain. ABT-102 decreased both spontaneous pain behaviors and those evoked by thermal and mechanical stimuli in these models. Moreover, we have found that repeated administration of ABT-102 for 5-12 days increased its analgesic activity in models of post-operative, osteoarthritic, and bone cancer pain without an associated accumulation of ABT-102 concentration in plasma or brain. Similar effects were also observed with a structurally distinct TRPV1 antagonist, A-993610. Although a single dose of ABT-102 produced a self-limiting increase in core body temperature that remained in the normal range, the hyperthermic effects of ABT-102 effectively tolerated following twice-daily dosing for 2 days. Therefore, the present data demonstrate that, following repeated administration, the analgesic activity of TRPV1 receptor antagonists is enhanced, while the associated hyperthermic effects are attenuated. The analgesic efficacy of ABT-102 supports its advancement into clinical studies.


Assuntos
Analgésicos/administração & dosagem , Febre/tratamento farmacológico , Indazóis/administração & dosagem , Limiar da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Canais de Cátion TRPV/metabolismo , Ureia/análogos & derivados , Animais , Temperatura Corporal/efeitos dos fármacos , Neoplasias Ósseas/complicações , Cálcio/metabolismo , Modelos Animais de Doenças , Interações Medicamentosas , Febre/induzido quimicamente , Inflamação/complicações , Masculino , Camundongos , Camundongos Endogâmicos C3H , Atividade Motora/efeitos dos fármacos , Osteoartrite/complicações , Dor/etiologia , Medição da Dor , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/antagonistas & inibidores , Ureia/administração & dosagem
8.
J Pharmacol Exp Ther ; 323(1): 217-26, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17636005

RESUMO

Calcium-sensing receptor (CaR) activation decreases serum parathyroid hormone (PTH) and Ca2+ and, despite long-term reductions in mean arterial blood pressure (MAP), may produce acute hypertension in rats, an effect we hypothesized was mediated by constriction of multiple vascular beds. Rats were subjected to 5/6 nephrectomy (NX) or no surgery (Normal); at 7 to 8 weeks, uremia animals were anesthetized and instrumented to record MAP and regional blood flow (carotid, mesenteric, and hindlimb). Cinacalcet [N-(1-naphthalen-1-ylethyl)-3-[3-(trifluoromethyl)phenyl]-propan-1-amine; 1, 3, and 10 mg/kg; 30 min/dose] was infused over 90 min. In NX rats, cinacalcet dose-dependently decreased ionized calcium (iCa2+), elicited a 90% reduction in PTH, and produced dose-dependent self-limiting increases in MAP (from 119 +/- 6 to 129 +/- 5, 142 +/- 4, and 145 +/- 3 mm Hg at the end of each infusion). At 1 mg/kg, carotid vascular resistance (CVR) and mesenteric vascular resistance (MVR) increased to 16 +/- 6 and 18 +/- 6% above baseline, respectively. Hindlimb vascular resistance (HVR) also trended upward (13 +/- 8%). At 3 mg/kg, increases in CVR (38 +/- 10%), MVR (40 +/- 8%), and HVR (39 +/- 14%) were exacerbated; at 10 mg/kg, values remained at or near these levels. The effects of cinacalcet in Normal rats were similar to NX and were attenuated by ganglionic blockade with hexamethonium at low doses but remained significantly elevated at higher doses. Thus, CaR activation acutely increases MAP in uremic and nonuremic rats, responses that occur in parallel to vasoconstriction in multiple vascular beds through both a central and peripheral mechanism of action. Moreover, subsequent mechanistic studies suggest that increases in MAP produced by cinacalcet may be mediated by reduced tonic NO synthase-dependent NO production subsequent to reductions in blood iCa2+.


Assuntos
Vias Aferentes/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Vias Eferentes/fisiologia , Tono Muscular/efeitos dos fármacos , Músculo Liso Vascular , Naftalenos/farmacologia , Receptores de Detecção de Cálcio/metabolismo , Uremia , Animais , Cálcio/sangue , Cinacalcete , Frequência Cardíaca/efeitos dos fármacos , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/inervação , Hormônio Paratireóideo/sangue , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Uremia/tratamento farmacológico , Uremia/metabolismo , Uremia/fisiopatologia , Resistência Vascular/efeitos dos fármacos
9.
J Med Chem ; 49(22): 6439-42, 2006 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17064063

RESUMO

Dipeptidyl peptidase IV (DPP4) deactivates glucose-regulating hormones such as GLP-1 and GIP, thus, DPP4 inhibition has become a useful therapy for type 2 diabetes. Optimization of the high-throughput screening lead 6 led to the discovery of 25 (ABT-341), a highly potent, selective, and orally bioavailable DPP4 inhibitor. When dosed orally, 25 dose-dependently reduced glucose excursion in ZDF rats. Amide 25 is safe in a battery of in vitro and in vivo tests and may represent a new therapeutic agent for the treatment of type 2 diabetes.


Assuntos
Compostos de Bifenilo/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptidil Peptidase 4/metabolismo , Hipoglicemiantes/farmacologia , Inibidores de Serina Proteinase/farmacologia , Triazóis/farmacologia , Animais , Compostos de Bifenilo/síntese química , Compostos de Bifenilo/farmacocinética , Cicloexenos/química , Diabetes Mellitus Tipo 2/genética , Relação Dose-Resposta a Droga , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Feminino , Hipoglicemiantes/síntese química , Hipoglicemiantes/farmacocinética , Modelos Moleculares , Ratos , Ratos Zucker , Inibidores de Serina Proteinase/síntese química , Inibidores de Serina Proteinase/farmacocinética , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/farmacocinética , Difração de Raios X
10.
J Med Chem ; 49(21): 6416-20, 2006 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17034148

RESUMO

Dipeptidyl peptidase-IV (DPP-IV) inhibitors are poised to be the next major drug class for the treatment of type 2 diabetes. Structure-activity studies of substitutions at the C5 position of the 2-cyanopyrrolidide warhead led to the discovery of potent inhibitors of DPP-IV that lack activity against DPP8 and DPP9. Further modification led to an extremely potent (Ki(DPP)(-)(IV) = 1.0 nM) and selective (Ki(DPP8) > 30 microM; Ki(DPP9) > 30 microM) clinical candidate, ABT-279, that is orally available, efficacious, and remarkably safe in preclinical safety studies.


Assuntos
Inibidores de Adenosina Desaminase , Inibidores da Dipeptidil Peptidase IV , Glicoproteínas/antagonistas & inibidores , Hipoglicemiantes/síntese química , Piridinas/síntese química , Pirrolidinas/síntese química , Adenosina Desaminase/química , Administração Oral , Animais , Sítios de Ligação , Células CACO-2 , Cristalografia por Raios X , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptidil Peptidase 4/química , Cães , Feminino , Intolerância à Glucose/tratamento farmacológico , Glicoproteínas/química , Humanos , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/farmacologia , Macaca fascicularis , Modelos Moleculares , Estrutura Molecular , Piridinas/farmacocinética , Piridinas/farmacologia , Pirrolidinas/farmacocinética , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Estereoisomerismo , Relação Estrutura-Atividade
11.
Curr Opin Investig Drugs ; 7(3): 206-13, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16555680

RESUMO

Vitamin D3 is modified by vitamin D3 25-hydroxylase in the liver, and by 25-hydroxyvitamin D3 1alpha-hydroxylase (CYP27B1) in the kidney, to form the active metabolite 1alpha,25-dihydroxyvitamin D3. Several vitamin D receptor (VDR) activators, including paricalcitol and calcitriol, are currently available for the treatment of hyperparathyroidism secondary to chronic kidney disease (CKD). CKD patients encounter a much higher risk of cardiovascular disease than do members of the general public, and recent clinical observations have shown that VDR activator therapy provides survival benefit for CKD patients in the rank order of paricalcitol > calcitriol > no VDR activator therapy, independent of parathyroid hormone, phosphorus and calcium. One possible explanation for this observation is that VDR activators exert a positive impact on cardiovascular functions. Studies in animals with disrupted genes involved in the vitamin D signaling pathway have provided some interesting data. For example, in mice lacking VDR or CYP27B1, it was found that in addition to the expected phenotype (hypocalcemia, secondary hyperparathyroidism and osteomalacia), expression of renin or atrial natriuretic peptide was elevated. The mice also developed hypertension and cardiac hypertrophy. Gene expression profiling studies have revealed that VDR may play a role in regulating smooth-muscle-cell (SMC) proliferation, thrombosis, fibrinolysis and vessel relaxation. Paricalcitol and calcitriol are equally potent at suppressing plasminogen activator inhibitor-1 synthesis and inhibiting cellular proliferation in human coronary artery SMCs. The effect of VDR activators on the modulation of renin expression and vascular functions may be factors that contribute to reduced mortality and morbidity risk in VDR-activator-treated CKD patients. In this review, we discuss recent preclinical and clinical data regarding the role of VDR and its ligands in the cardiovascular system.


Assuntos
Calcitriol/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Ergocalciferóis/uso terapêutico , Falência Renal Crônica/tratamento farmacológico , Receptores de Calcitriol/metabolismo , Ensaios Clínicos como Assunto , Humanos , Modelos Biológicos
12.
J Med Chem ; 49(7): 2339-52, 2006 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-16570930

RESUMO

An inactin-anesthetized rat cardiovascular (CV) assay was employed in a screening mode to triage multiple classes of melanin-concentrating hormone receptor 1 (MCHr1) antagonists. Lead identification was based on a compound profile producing high drug concentration in both plasma (>40 microM) and brain (>20 microg/g) with <15% change in cardiovascular endpoints. As a result of these stringent requirements, lead optimization activities on multiple classes of MCHr1 antagonists were terminated. After providing evidence that the cardiovascular liabilities were not a function of MCHr1 antagonism, continued screening identified the chromone-substituted aminopiperidine amides as a class of MCHr1 antagonists that demonstrated a safe cardiovascular profile at high drug concentrations in both plasma and brain. The high incidence of adverse cardiovascular effects associated with an array of MCHr1 antagonists of significant chemical diversity, combined with the stringent safety requirements for antiobesity drugs, highlight the importance of incorporating cardiovascular safety assessment early in the lead selection process.


Assuntos
Fármacos Antiobesidade/síntese química , Sistema Cardiovascular/efeitos dos fármacos , Cromonas/síntese química , Piperidinas/síntese química , Receptores de Somatostatina/antagonistas & inibidores , Animais , Fármacos Antiobesidade/efeitos adversos , Fármacos Antiobesidade/sangue , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular Tumoral , Cromonas/efeitos adversos , Cromonas/sangue , Cães , Indazóis/efeitos adversos , Indazóis/sangue , Indazóis/síntese química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Piperidinas/efeitos adversos , Piperidinas/sangue , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Distribuição Tecidual
13.
J Cardiovasc Pharmacol ; 46(2): 232-40, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16044036

RESUMO

The pharmacological profile of the novel dihydropyridine K channel opener (KCO), (9R)-9-(3-iodo-4-methylphenyl)-5,9-dihydro-3H-furo[3,4-b]pyrano[4,3-e]pyridine-1,8(4H,7H)-dione (A-325100), is described in numerous in vitro assays. Furthermore, the cardiovascular effects of A-325100 are characterized in both the anesthetized and conscious dog. In vitro, A-325100 selectively activated KATP currents and potently relaxed vascular smooth muscle (IC50 between 7.69x10 M and 7.78x10 M), an effect that was abolished by glyburide. Moreover, A-325100 did not interact with L-type Ca2+ channels at concentrations up to 30 microM. In anesthetized dogs A-325100 produced a dose-dependent reduction in systemic vascular resistance and mean arterial pressure concomitant with dose-dependent increases in dP/dtmax and heart rate. In conscious telemetry-instrumented dogs oral administration of A-325100 produced a similar response profile, including dose-dependent reductions in MAP and increases in heart rate and dP/dtmax. When concentration-dependent changes in MAP, heart rate, and dP/dtmax were compared relative to circulating plasma concentrations, A-325100 produced similar effects in both the anesthetized and conscious dog. In conclusion, the present study provides the first pharmacological description of the novel and selective tricyclic dihydropyridine KCO, A-325100. When studied in vivo, A-325100 produced similar concentration-dependent cardiovascular effects in both models consistent with its mode of action and independent of route of administration. Thus, these data demonstrate that the hemodynamic effects of vasoactive compounds, such as KCOs, can be effectively profiled in both the conscious and anesthetized dog.


Assuntos
Di-Hidropiridinas/farmacologia , Hemodinâmica/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio/metabolismo , Pironas/farmacologia , Trifosfato de Adenosina/metabolismo , Anestesia , Animais , Aorta Torácica/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Di-Hidropiridinas/sangue , Cães , Relação Dose-Resposta a Droga , Cobaias , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Ligantes , Masculino , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Técnicas de Patch-Clamp , Veia Porta/efeitos dos fármacos , Pironas/sangue , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/fisiologia
14.
Clin Exp Pharmacol Physiol ; 32(12): 1020-6, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16445566

RESUMO

Iron-deficiency anaemia, a complication of end-stage renal disease (ESRD), is often treated with parenteral iron therapies that have been shown to produce dose-limiting hypotension in patients. ABT-870 (iron-(III)-hydroxide-oligosaccharide) is comprised of elemental iron complexed with oligosaccharide, a composition that we hypothesised would allow the hypotensive effects of parenteral iron therapy to be overcome, thus allowing a rapid rate of infusion to be well tolerated. Mean arterial pressure (MAP) and heart rate (HR) were monitored in anaesthetized dogs following the infusion of ABT-870 and iron sucrose administered at doses of 7.1 and 21.3 mg/kg using a rapid 30 s infusion. ABT-870 and iron sucrose were also monitored at doses of 7.1, 21.3 and 50 mg/kg administered over a 10 min period. Sodium ferric gluconate complex (SFGC) was administered in an identical fashion at doses of 12.5 and 31.2 mg/kg. A 30 s rapid infusion of ABT-870 at doses of 7.1 and 14.3 mg/kg or a 10 min infusion of ABT-870 at doses of 7.1 and 21.3 mg/kg produced little effect on MAP and HR. Infusion of the highest dose of ABT-870 (50 mg/kg) produced no consistent hypotension, but did produce an increase in HR (maximal increase 35 +/- 9 b.p.m.), an effect that lasted only 15 min. A 30 s rapid infusion of iron sucrose at 7.1 mg/kg produced modest increases in MAP and HR (5 +/- 1 mmHg and 5 +/- 2 b.p.m., respectively). However, rapid infusion of iron sucrose at 14.3 mg/kg produced hypotension (to -8 +/- 1 mmHg below baseline) and exerted variable, biphasic effects on HR ranging from -16 to +50 b.p.m. Although 10 min infusion of iron sucrose at 7.1 mg/kg exerted little effect on MAP and HR, at doses of 21.3 and 50 mg/kg iron sucrose elicited a profound dose-dependent decrease in MAP (-34 +/- 11 and -83 +/- 5 mmHg, respectively) and a pronounced increase in HR ranging from 32 to 49 b.p.m. above baseline. A 10 min infusion of SFGC at doses of 12.5 and 31.2 mg/kg produced a dose-dependent decrease in MAP (-28 +/- 18 and -67 +/- 12 mmHg below baseline) and a marked increase in HR (26 +/- 11 and 94 +/- 15 b.p.m. above baseline). In conclusion, unlike iron sucrose and SFGC, high doses of ABT-870 failed to exert consistent hypotensive effects. These data demonstrate that ABT-870 may have a substantial therapeutic window and considerable clinical potential for iron-replacement therapy.


Assuntos
Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Compostos Férricos/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Hematínicos/farmacologia , Compostos de Ferro/farmacologia , Oligossacarídeos/farmacologia , Animais , Dextranos , Cães , Compostos Férricos/administração & dosagem , Compostos Férricos/química , Infusões Intravenosas , Masculino , Oligossacarídeos/administração & dosagem
15.
J Cardiovasc Pharmacol ; 44(2): 137-47, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15243293

RESUMO

The therapeutic utility of KATP channel opening agents (KCOs) in the treatment of overactive bladder may be limited by hypotension as a result of insufficient selectivity in vivo for bladder versus vasculature smooth muscle. Recently, we demonstrated that the putative uroselective KCOs, A-278637, ZD-6169, and WAY-133537 suppress unstable bladder contraction in an in vivo pre-clinical pig model of detrusor instability secondary to partial outlet obstruction. In the present study in the anesthetized dog we targeted plasma concentrations 3-, 10-, and 30-fold above a common index of in vivo efficacy (EC35) for suppression of unstable bladder contraction in pigs, to provide a comprehensive cardiovascular profile of these compounds. When compared at similar multiples of efficacy, dose-dependent reductions in SVR were greater in ZD-6169 and WAY-133537-treated animals versus A-278637. A-278637, unlike ZD-6169 or WAY-133537, produced no effect on MAP at concentrations 10-fold above the EC35. At concentrations 30-fold above the EC35, MAP in A-278637-treated animals was reduced -11% from baseline versus -24% and -42% for ZD-6169 and WAY-133537. Accordingly, at plasma concentrations approximately 30-fold above the EC35 reflex-mediated increases in HR were modest for A-278637-treated animals (15% above baseline) versus ZD-6169 (22%) or WAY-133537 (35%). Increases in both dP/dt and cardiac output occurred at lower therapeutic multiples and were greater in magnitude for animals treated with WAY-133537 (66% and 64% above baseline, respectively, 60 minutes into compound infusion) and ZD-6169 (10% and 13%) versus A-278637 (-2% and 6%). Thus, A-278637 exerted lesser effects on cardiovascular function at equivalent multiples of the EC35 than either ZD-6169 or WAY-133537. These data suggest that A-278637 possesses a greater functional selectivity for urinary bladder versus vascular smooth muscle in vivo and that A-278637 may exhibit a more favorable therapeutic index than either ZD-6169 or WAY-133537.


Assuntos
Amidas/farmacologia , Benzofenonas/farmacologia , Óxidos S-Cíclicos/farmacologia , Ciclobutanos/farmacologia , Nifedipino/farmacologia , Nitrilas/farmacologia , Canais de Potássio/fisiologia , Quinolonas/farmacologia , Trifosfato de Adenosina/química , Amidas/sangue , Animais , Benzofenonas/sangue , Pressão Sanguínea/efeitos dos fármacos , Óxidos S-Cíclicos/sangue , Ciclobutanos/sangue , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Eletrocardiografia/métodos , Frequência Cardíaca/efeitos dos fármacos , Hipotensão/induzido quimicamente , Infusões Intravenosas , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Nifedipino/sangue , Nitrilas/sangue , Veículos Farmacêuticos/administração & dosagem , Veículos Farmacêuticos/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Canais de Potássio/efeitos dos fármacos , Quinolonas/sangue , Taquicardia/induzido quimicamente , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/fisiologia , Incontinência Urinária/tratamento farmacológico , Incontinência Urinária/fisiopatologia , Resistência Vascular/efeitos dos fármacos
16.
Circulation ; 107(14): 1912-6, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12668514

RESUMO

BACKGROUND: Embryonic stem (ES) cells are capable of self-renewal and differentiation into cellular derivatives of all 3 germ layers. In appropriate culture conditions, ES cells can differentiate into specialized cells, including cardiac myocytes, but the efficiency is typically low and the process is incompletely understood. METHODS AND RESULTS: We evaluated a chemical library for its potential to induce cardiac differentiation of ES cells in the absence of embryoid body formation. Using ES cells stably transfected with cardiac-specific alpha-cardiac myosin heavy chain (MHC) promoter-driven enhanced green fluorescent protein (EGFP), 880 compounds approved for human use were screened for their ability to induce cardiac differentiation. Treatment with ascorbic acid, also known as vitamin C, markedly increased the number of EGFP-positive cells, which displayed spontaneous and rhythmic contractile activity and stained positively for sarcomeric myosin and alpha-actinin. Furthermore, ascorbic acid induced the expression of cardiac genes, including GATA4, alpha-MHC, and beta-MHC in untransfected ES cells in a developmentally controlled manner. This effect of ascorbic acid on cardiac differentiation was not mimicked by the other antioxidants such as N-acetylcysteine, Tiron, or vitamin E. CONCLUSIONS: Ascorbic acid induces cardiac differentiation in ES cells. This study demonstrates the potential for chemically modifying the cardiac differentiation program of ES cells.


Assuntos
Ácido Ascórbico/farmacologia , Embrião de Mamíferos/citologia , Embrião não Mamífero , Miócitos Cardíacos/citologia , Células-Tronco/fisiologia , Actinina/análise , Animais , Antioxidantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Miócitos Cardíacos/química , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/genética , Miosinas/análise , RNA Mensageiro/biossíntese , Células-Tronco/efeitos dos fármacos
17.
FASEB J ; 16(11): 1468-70, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12205051

RESUMO

Opioids and volatile anesthetics produce marked cardioprotective effects against myocardial infarction via the activation of ATP-sensitive potassium (K(ATP)) channels, however, the effect of combined treatment with both drugs is unknown. We examined the hypothesis that opioids and volatile anesthetics potentiate cardiac K(ATP) channel opening, thereby enhancing cardioprotection. Rats were treated with the delta opioid agonists, TAN-67 or BW373U86, or isoflurane, together or alone with and without diazoxide, a mitochondrial K(ATP) channel opener. Glibenclamide, a non-selective K(ATP) channel blocker, was used to further characterize the signaling mechanism involved. Myocardial infarct size (IS) was determined by tetrazolium staining and was expressed as a percent of the area at risk (AAR). High doses of TAN-67 (10 mg/kg), diazoxide (10 mg/kg), and isoflurane (1 MAC) produced a significant reduction in IS compared with the control group (30+/-3%, 36+/-5%, and 42+/-2 vs. 58+/-2%, respectively), whereas lower doses of the drugs had no effect except for the low dose of isoflurane (0.5 MAC). The combination of TAN-67 and diazoxide or isoflurane and diazoxide resulted in a marked reduction in IS compared with controls in the presence of high (9+/-3% and 14+/-3%) and low (17+/-7% and 31+/-7%) dose combinations, respectively. The combination of TAN-67 or BW373U86 and isoflurane also caused a striking reduction in IS/AAR (16+/-7% and 7+/-2%, respectively). To date, this is the first demonstration that opioids and volatile anesthetics work in conjunction to confer protection against myocardial infarction through potentiation of cardiac K(ATP) channel opening.


Assuntos
Anestésicos Inalatórios/farmacologia , Cardiotônicos/farmacologia , Isoflurano/farmacologia , Infarto do Miocárdio/prevenção & controle , Canais de Potássio/metabolismo , Receptores Opioides delta/agonistas , Trifosfato de Adenosina/metabolismo , Animais , Benzamidas/farmacologia , Diazóxido/farmacologia , Sinergismo Farmacológico , Modelos Cardiovasculares , Miocárdio/metabolismo , Entorpecentes/farmacologia , Piperazinas/farmacologia , Quinolinas/farmacologia , Ratos
18.
Cardiovasc Res ; 55(3): 520-5, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12160948

RESUMO

This review focuses on target receptors that have been shown to have the potential to mimic the cardioprotective effect of ischemic preconditioning (IPC). There is an abundance of information concerning the intracellular mechanisms and membrane-bound receptors responsible for IPC. Important intracellular mediators of this cardioprotection likely reside in the activation of multiple kinase cascades. The major players in IPC are thought to include protein kinase C, tyrosine kinases, and members of the mitogen-activated protein kinase signaling family and these topics will be covered in more detail in other papers of this focused issue. However, many of these kinase-mediated mechanisms are triggered by the activation of transmembrane spanning receptors, some of which may be manipulated therapeutically to induce cardioprotection in humans with unstable angina or who are at risk for myocardial infarction. In this review, we will discuss the evidence supporting the possibility of manipulating several of these G protein-coupled receptors as potential therapeutic targets. Stimulation of numerous receptors has been targeted as possible triggers for IPC. Some of those that have been identified include A(1) adenosine, alpha(1) adrenergic, M(2) muscarinic, B(2) bradykinin, delta(1) opioid, AT(1) angiotensin, and endothelin-1 receptors. In general, these receptors are thought to couple to inhibitory G proteins. In this review, we will focus on the most likely therapeutic candidates for cardioprotection, namely adenosine, opioid, and bradykinin receptors since selective agonists and antagonists, either alone or in combination, have most often been shown to mimic or block IPC in numerous animal models and man, respectively. This is not meant to completely rule out other receptors since it is clear that IPC is a phenomenon with multiple pathways that appear to be responsible for the cardioprotection observed.


Assuntos
Antagonistas dos Receptores da Bradicinina , Precondicionamento Isquêmico Miocárdico , Isquemia Miocárdica/tratamento farmacológico , Antagonistas de Entorpecentes , Antagonistas de Receptores Purinérgicos P1 , Adenosina/metabolismo , Animais , Bradicinina/metabolismo , Humanos , Isquemia Miocárdica/metabolismo , Entorpecentes/metabolismo , Receptores da Bradicinina/metabolismo , Receptores Opioides/metabolismo , Receptores Purinérgicos P1/metabolismo
19.
Basic Res Cardiol ; 97(1): 47-54, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11998977

RESUMO

We have previously demonstrated that cardioprotection induced by the infusion of a selective delta1-opioid agonist is mediated by the specific translocation of PKC-delta to the mitochondria in in vivo rat hearts and via opening of the mitochondrial KATP channel. Ischemic preconditioning (IPC) is also thought to involve the translocation of specific isoforms of PKC and KATP channel activation. Therefore, we utilized the PKC-delta selective antagonist, rottlerin, to assess the effect of inhibition of this isozyme on cardioprotection induced by one-cycle of IPC prior to 30 minutes of ischemia and 2 hours of reperfusion. Infarct size (IS) was determined by tetrazolium chloride staining and expressed as a percent of the area at risk (AAR). Non-preconditioned control animals had an IS/AAR of 59.7 +/- 1.6. IPC significantly reduced the extent of myocardial infarction (6.3 +/- 1.4). Rottlerin, 0.3 mg/kg, did not alter IS/AAR in control animals (55.0 +/- 5.6), and had no significant effect on IS/AAR in preconditioned animals (14.4 +/- 3.8). Additionally, we demonstrated, using a luciferase-based assay to determine the rate of ATP synthesis and state of mitochondrial bioenergetics, that IPC preserves ATP synthesis in the ischemic myocardium and that this preservation is attenuated by the isoform non-selective PKC inhibitor, chelerythrine, but not by the delta-selective antagonist, rottlerin. These data suggest that PKC-delta does not play an important role in IPC and that differences in isoform importance are evident during pharmacological versus ischemia-induced preconditioning.


Assuntos
Trifosfato de Adenosina/biossíntese , Precondicionamento Isquêmico Miocárdico , Isoenzimas/antagonistas & inibidores , Mitocôndrias Cardíacas/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Proteína Quinase C/antagonistas & inibidores , Acetofenonas/farmacologia , Alcaloides , Animais , Benzofenantridinas , Benzopiranos/farmacologia , Inibidores Enzimáticos/farmacologia , Hemodinâmica , Imuno-Histoquímica , Isoenzimas/metabolismo , Masculino , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/enzimologia , Infarto do Miocárdio/enzimologia , Fenantridinas/farmacologia , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA