Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 356(2): 333-40, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26585572

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is prevalent and often accompanied by metabolic syndrome. Current treatment options are limited. Here, we test the hypothesis that combined A1/A2B adenosine receptor blockade is beneficial in obese ZSF1 rats, an animal model of HFpEF with metabolic syndrome. The combined A1/A2B receptor antagonist 3-[4-(2,6-dioxo-1,3-dipropyl-7H-purin-8-yl)-1-bicyclo[2.2.2]octanyl]propanoic acid (BG9928) was administered orally (10 mg/kg/day) to obese ZSF1 rats (n = 10) for 24 weeks (from 20 to 44 weeks of age). Untreated ZSF1 rats (n = 9) served as controls. After 24 weeks of administration, BG9928 significantly lowered plasma triglycerides (in mg/dl: control group, 4351 ± 550; BG9928 group, 2900 ± 551) without adversely affecting plasma cholesterol or activating renin release. BG9928 significantly decreased 24-hour urinary glucose excretion (in mg/kg/day: control group, 823 ± 179; BG9928 group, 196 ± 80) and improved oral glucose tolerance, polydipsia, and polyuria. BG9928 significantly augmented left ventricular diastolic function in association with a reduction in cardiac vasculitis and cardiac necrosis. BG9928 significantly reduced 24-hour urinary protein excretion (in mg/kg/day: control group, 1702 ± 263; BG9928 group, 1076 ± 238), and this was associated with a reduction in focal segmental glomerulosclerosis, tubular atrophy, tubular dilation, and deposition of proteinaceous material in the tubules. These findings show that, in a model of HFpEF with metabolic syndrome, A1/A2B receptor inhibition improves hyperlipidemia, exerts antidiabetic actions, reduces HFpEF, improves cardiac histopathology, and affords renal protection. We conclude that chronic administration of combined A1/A2B receptor antagonists could be beneficial in patients with HFpEF, in particular those with comorbidities such as obesity, diabetes, and dyslipidemias.


Subject(s)
Adenosine A1 Receptor Antagonists/administration & dosage , Adenosine A2 Receptor Antagonists/administration & dosage , Heart Failure/drug therapy , Kidney/physiology , Stroke Volume/drug effects , Xanthines/administration & dosage , Animals , Heart Failure/metabolism , Kidney/drug effects , Male , Rats , Rats, Zucker , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2B/metabolism , Stroke Volume/physiology , Treatment Outcome
2.
Expert Opin Drug Discov ; 7(12): 1129-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23095011

ABSTRACT

INTRODUCTION: Kinins are bioactive peptide hormones that exert biological effects by activating two types of G protein-coupled receptors namely, B(1) (B(1)R) and B(2) (B(2)R). These modulate normal physiological cellular functions, inflammatory disorders and carcinogenesis. New and novel kinin receptor antagonists have been synthesized and their efficacy evaluated. AREAS COVERED: The authors provide a comprehensive review on the cellular and molecular biology of kinins and their receptors is delineated along with evolution and discovery of selective peptide and non-peptide antagonists. The authors describe the in vitro and in vivo methods used to understand the relative functional roles of B(1)R and B(2)R in physiology and pathohysiology. Furthermore, the authors translate the evaluation of kinin antagonists in selected preclinical models and associated clinical indications. Literature was surveyed from original publications, standard sources, SciFinder, patent applications and clinical trials. EXPERT OPINION: The authors suggest that several key areas of functional biology need consideration, namely: re-evaluation, particularly in vivo, of the mechanism of action and relative functional roles of the B(1)R and B(2)R in physiology and acute and chronic disease in animals and man; need for improved animal models with increased use of humanized and human systems; development of fluorescent probes for use in vivo in animals and man using advanced imaging techniques; combination of kinin receptor antagonists and traditional chemotherapy for various cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Bradykinin B1 Receptor Antagonists , Bradykinin B2 Receptor Antagonists , Neoplasms/drug therapy , Animals , Drug Discovery , Humans , Kinins/physiology , Models, Animal
3.
J Cereb Blood Flow Metab ; 30(1): 130-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19773800

ABSTRACT

Inflammatory mechanisms are known to contribute to the pathophysiology of traumatic brain injury (TBI). Since bradykinin is one of the first mediators activated during inflammation, we investigated the role of bradykinin and its receptors in posttraumatic secondary brain damage. We subjected wild-type (WT), B(1)-, and B(2)-receptor-knockout mice to controlled cortical impact (CCI) and analyzed tissue bradykinin as well as kinin receptor mRNA and protein expression up to 48 h thereafter. Brain edema, contusion volume, and functional outcome were assessed 24 h and 7 days after CCI. Tissue bradykinin was maximally increased 2 h after trauma (P<0.01 versus sham). Kinin B(1) receptor mRNA was upregulated up to four-fold 24 h after CCI. Immunohistochemistry showed that B(1) and B(2) receptors were expressed in the brain and were significantly upregulated in the traumatic penumbra 1 to 24 h after CCI. B(2)R(-/-) mice had significantly less brain edema (-51% versus WT, 24 h; P<0.001), smaller contusion volumes ( approximately 50% versus WT 24 h and 7 d after CCI; P<0.05), and better functional outcome 7 days after TBI as compared with WT mice (P<0.05). The present results show that bradykinin and its B(2) receptors play a causal role for brain edema formation and cell death after TBI.


Subject(s)
Brain Injuries/pathology , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Animals , Bradykinin/metabolism , Brain Edema/pathology , Contusions/pathology , Immunohistochemistry , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Bradykinin B1/biosynthesis , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/biosynthesis , Receptor, Bradykinin B2/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Nephrol ; 30(6): 521-6, 2009.
Article in English | MEDLINE | ID: mdl-19828940

ABSTRACT

BACKGROUND/AIMS: Cisplatin (CIS) induces nephrotoxicity partly through renal vasoconstriction and decreased glomerular filtration effects thought to involve adenosine acting on adenosine A(1) receptors (A1Rs). We studied the effect of the orally active, A1R antagonist tonapofylline (BG9928) on biochemical measures of renal function in CIS-induced acute kidney injury (AKI) in rats. METHODS: Tonapofylline, 1 mg/kg b.i.d., p.o., was administered on days 0-1 or 0-6 to rats treated with CIS 5.5 mg/kg i.v. Prednisolone (PRED) 5 mg/kg s.c. (day 0) served as a positive control. Serum creatinine and urea nitrogen (BUN) were measured in serial blood samples taken over the 13-day study period. RESULTS: CIS produced significant elevations in creatinine, reduction in body weight and marked proximal tubular injury throughout the renal cortex and outer medulla. Tonapofylline, days 0-1 or 0-6 and PRED all produced sustained reductions in post-CIS serum creatinine and BUN levels compared with controls, improved body weight recovery and significant attenuation of CIS-induced kidney pathology scores. CONCLUSION: These data support the involvement of A1Rs in CIS-induced AKI in rats. Tonapofylline may be useful in the clinical setting for the prevention of kidney failure induced by nephrotoxic agents such as CIS.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Adenosine A1 Receptor Antagonists , Cisplatin/toxicity , Xanthines/pharmacology , Acute Kidney Injury/pathology , Animals , Antineoplastic Agents/toxicity , Blood Urea Nitrogen , Creatinine/blood , Cytoprotection , Drug Interactions , Female , Glucocorticoids/pharmacology , Kidney Function Tests , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Prednisolone/pharmacology , Rats , Rats, Sprague-Dawley
5.
Bioorg Med Chem Lett ; 15(3): 511-5, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15664803

ABSTRACT

A novel [1,2,4]triazolo[1,5-a]pyrazine core was synthesized and coupled with terminal acetylenes. The structure-activity relationship of the alkynes from this novel template was studied for their in vitro and in vivo adenosine A(2A) receptor antagonism. Selected compounds from this series were shown to have potent in vitro and in vivo activities against adenosine A(2A) receptor. Compound 12, in particular, was found to be orally active at 3mg/kg in both a mouse catalepsy model and a 6-hydroxydopamine-lesioned rat model.


Subject(s)
Adenosine A2 Receptor Antagonists , Pyrazines/chemical synthesis , Pyrazines/pharmacology , Administration, Oral , Alkynes/chemistry , Animals , Catalepsy/drug therapy , Cerebral Cortex , Disease Models, Animal , Mice , Oxidopamine , Parkinson Disease/drug therapy , Pyrazines/administration & dosage , Rats , Structure-Activity Relationship , Triazoles/chemical synthesis
6.
J Med Chem ; 47(25): 6218-29, 2004 Dec 02.
Article in English | MEDLINE | ID: mdl-15566292

ABSTRACT

A series of bicyclic piperazine derivatives of triazolotriazine and triazolopyrimidines was synthesized. Some of these analogues show high affinity and excellent selectivity for adenosine A(2a) receptor versus the adenosine A(1) receptor. Structure-activity-relationship (SAR) studies based on octahydropyrrolo[1,2-a]pyrazine and octahydropyrido[1,2-a]pyrazine with various capping groups are reported. Among these analogues, the most potent and selective A(2a) antagonist 26 h has a K(i) value of 0.2 nM and is 16 500-fold selective with respect to the A(1) receptor. Among a number of compounds tested, compounds 21a and 21c exhibited significantly improved metabolic stability. Compounds 21a, 21c, and 18a showed good oral efficacy in rodent catalepsy models of Parkinson's disease.


Subject(s)
Adenosine A2 Receptor Antagonists , Piperazines/chemical synthesis , Pyrimidines/chemical synthesis , Triazines/chemical synthesis , Triazoles/chemical synthesis , Administration, Oral , Animals , Catalepsy/drug therapy , Disease Models, Animal , Drug Stability , In Vitro Techniques , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Parkinson Disease/drug therapy , Piperazines/chemistry , Piperazines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
7.
J Pharmacol Exp Ther ; 308(3): 846-56, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14634049

ABSTRACT

A(1) adenosine receptor (AR) antagonists are effective diuretic agents that may be useful for treating fluid retention disorders including congestive heart failure. However, antagonism of A(1)ARs is potentially a concern when using these agents in patients with ischemic heart disease. To address this concern, the present study was designed to compare the actions of the A(1)AR antagonists CPX (1,3-dipropyl-8-cyclopentylxanthine), BG 9719 (1,3-dipropyl-8-[2-(5,6-epoxynorbornyl)]xanthine), and BG 9928 (1,3-dipropyl-8-[1-(4-propionate)-bicyclo-[2,2,2]octyl]xanthine) on acute myocardial ischemia/reperfusion injury and ischemic preconditioning (IPC) in an in vivo dog model of infarction. Barbital-anesthetized dogs were subjected to 60 min of left anterior descending coronary artery occlusion followed by 3 h of reperfusion, after which infarct size was assessed by staining with triphenyltetrazolium chloride. IPC was elicited by four 5-min occlusion/5-min reperfusion cycles produced 10 min before the 60-min occlusion. Multiple-cycle IPC produced a robust reduction ( approximately 65%) in infarct size; this effect of IPC on infarct size was not abrogated in dogs pretreated with any of the three AR antagonists. Surprisingly, in the absence of IPC, pretreatment with CPX or BG 9928 before occlusion or immediately before reperfusion resulted in significant reductions ( approximately 40-50%) in myocardial infarct size. However, treatment with an equivalent dose of BG 9719 had no similar effect. We conclude that the A(1)AR antagonists BG 9719, BG 9928, and CPX do not exacerbate cardiac injury and do not interfere with IPC induced by multiple ischemia/reperfusion cycles. We discuss the possibility that the cardioprotective actions of CPX and BG 9928 may be related to antagonism of A(2B)ARs.


Subject(s)
Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Xanthines/therapeutic use , Adenosine A1 Receptor Antagonists , Anesthesia , Animals , Disease Models, Animal , Dogs , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/complications , Radioligand Assay , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/metabolism
8.
Blood ; 102(13): 4464-71, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12933585

ABSTRACT

Interaction of very late antigen-4 (VLA-4) with its ligand vascular cell adhesion molecule-1 (VCAM-1) is required for central nervous system (CNS) migration of encephalitogenic T cells in relapsing experimental autoimmune encephalomyelitis (R-EAE). Anti-VLA-4 monoclonal antibody (mAb) treatment prior to EAE onset inhibits disease induction; however, treatment initiated after the appearance of clinical symptoms increases relapse rates, augments Th1 responses, and enhances epitope spreading perhaps due to the activation of costimulatory signals. To negate the potential costimulatory activity of intact anti-VLA-4, we examined the ability of BIO 5192, a small-molecule VLA-4 antagonist, to regulate active proteolipid protein 139-151 (PLP139-151)-induced R-EAE. BIO 5192 administered one week after peptide priming (ie, before clinical disease onset) delayed the clinical disease onset but led to severe disease exacerbation upon treatment removal. BIO 5192 treatment initiated during disease remission moderately enhanced clinical disease while mice were on treatment and also resulted in posttreatment exacerbation. Interestingly, BIO 5192 treatment begun at the peak of acute disease accelerated entrance into disease remission and inhibited relapses, but treatment removal again exacerbated disease. Enhanced disease was caused by the release of encephalitogenic cells from the periphery and the rapid accumulation of T cells in the CNS. Collectively, these results further demonstrate the complexity of VLA-4/VCAM interactions, particularly in a relapsing-remitting autoimmune disease.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Integrin alpha4beta1/antagonists & inhibitors , Oligopeptides/therapeutic use , Phenylurea Compounds/therapeutic use , Th1 Cells/pathology , Amino Acid Sequence , Animals , Blood-Brain Barrier/immunology , Cell Adhesion/drug effects , Central Nervous System/immunology , Disease Models, Animal , Disease Progression , Drug Administration Schedule , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Integrin alpha4beta1/physiology , Mice , Mice, Inbred Strains , Molecular Sequence Data , Multiple Sclerosis, Relapsing-Remitting , Myelin Proteolipid Protein/immunology , Oligopeptides/administration & dosage , Oligopeptides/toxicity , Peptide Fragments/immunology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/toxicity , Recurrence , Th1 Cells/immunology , Vascular Cell Adhesion Molecule-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL