Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 24(17)2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37686032

ABSTRACT

Chronic kidney disease (CKD) progression is associated with persisting oxidative stress, which impairs the NO-sGC-cGMP signaling cascade through the formation of oxidized and heme-free apo-sGC that cannot be activated by NO. Runcaciguat (BAY 1101042) is a novel, potent, and selective sGC activator that binds and activates oxidized and heme-free sGC and thereby restores NO-sGC-cGMP signaling under oxidative stress. Therefore, runcaciguat might represent a very effective treatment option for CKD/DKD. The potential kidney-protective effects of runcaciguat were investigated in ZSF1 rats as a model of CKD/DKD, characterized by hypertension, hyperglycemia, obesity, and insulin resistance. ZSF1 rats were treated daily orally for up to 12 weeks with runcaciguat (1, 3, 10 mg/kg/bid) or placebo. The study endpoints were proteinuria, kidney histopathology, plasma, urinary biomarkers of kidney damage, and gene expression profiling to gain information about relevant pathways affected by runcaciguat. Furthermore, oxidative stress was compared in the ZSF1 rat kidney with kidney samples from DKD patients. Within the duration of the 12-week treatment study, kidney function was significantly decreased in obese ZSF1 rats, indicated by a 20-fold increase in proteinuria, compared to lean ZSF1 rats. Runcaciguat dose-dependently and significantly attenuated the development of proteinuria in ZSF1 rats with reduced uPCR at the end of the study by -19%, -54%, and -70% at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo treatment. Additionally, average blood glucose levels measured as HbA1C, triglycerides, and cholesterol were increased by five times, twenty times, and four times, respectively, in obese ZSF1 compared to lean rats. In obese ZSF1 rats, runcaciguat reduced HbA1c levels by -8%, -34%, and -76%, triglycerides by -42%, -55%, and -71%, and cholesterol by -16%, -17%, and -34%, at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo. Concomitantly, runcaciguat also reduced kidney weights, morphological kidney damage, and urinary and plasma biomarkers of kidney damage. Beneficial effects were accompanied by changes in gene expression that indicate reduced fibrosis and inflammation and suggest improved endothelial stabilization. In summary, the sGC activator runcaciguat significantly prevented a decline in kidney function in a DKD rat model that mimics common comorbidities and conditions of oxidative stress of CKD patients. Thus, runcaciguat represents a promising treatment option for CKD patients, which is in line with recent phase 2 clinical study data, where runcaciguat showed promising efficacy in CKD patients (NCT04507061).


Subject(s)
Kidney , Renal Insufficiency, Chronic , Animals , Rats , Cyclic GMP , Glycated Hemoglobin , Heme , Obesity , Proteinuria , Renal Insufficiency, Chronic/drug therapy , Clinical Trials, Phase II as Topic
3.
Toxicol Sci ; 187(1): 35-50, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35244176

ABSTRACT

Micro-RNAs (miRNAs) are regulators of gene expression and play an important role in physiological homeostasis and disease. In biofluids, miRNAs can be found in protein complexes or in extracellular vesicles (EVs). Altered urinary miRNAs are reported as potential biomarkers for chronic kidney disease (CKD). In this context, we compared established urinary protein biomarkers for kidney injury with urinary miRNA profiles in obese ZSF1 and hypertensive renin transgenic rats. Additionally, the benefit of urinary EV enrichment was investigated in vivo and the potential association of urinary miRNAs with renal fibrosis in vitro. Kidney damage in both rat models was confirmed by histopathology, proteinuria, and increased levels of urinary protein biomarkers. In total, 290 miRNAs were elevated in obese ZSF1 rats compared with lean controls, whereas 38 miRNAs were altered in obese ZSF1 rats during 14-26 weeks of age. These 38 miRNAs correlated better with disease progression than established urinary protein biomarkers. MiRNAs increased in obese ZSF1 rats were associated with renal inflammation, fibrosis, and glomerular injury. Eight miRNAs were also changed in urinary EVs of renin transgenic rats, including one which might play a role in endothelial dysfunction. EV enrichment increased the number and detection level of several miRNAs implicated in renal fibrosis in vitro and in vivo. Our results show the benefit of EV enrichment for miRNA detection and the potential of total urine and urinary EV-associated miRNAs as biomarkers of altered kidney physiology, renal fibrosis and glomerular injury, and disease progression in hypertension and obesity-induced CKD.


Subject(s)
Extracellular Vesicles , Hypertension , MicroRNAs , Renal Insufficiency, Chronic , Animals , Biomarkers/metabolism , Disease Progression , Extracellular Vesicles/metabolism , Female , Fibrosis , Humans , Hypertension/metabolism , Kidney/metabolism , Male , MicroRNAs/genetics , Obesity/metabolism , Rats , Renin/metabolism
4.
Br J Pharmacol ; 179(11): 2476-2489, 2022 06.
Article in English | MEDLINE | ID: mdl-34096053

ABSTRACT

BACKGROUND AND PURPOSE: Generation of cGMP via NO-sensitive soluble guanylyl cyclase (sGC) has been implicated in the regulation of renal functions. Chronic kidney disease (CKD) is associated with decreased NO bioavailability, increased oxidative stress and oxidation of sGC to its haem-free form, apo-sGC. Apo-sGC cannot be activated by NO, resulting in impaired cGMP signalling that is associated with chronic kidney disease progression. We hypothesised that sGC activators, which activate apo-sGC independently of NO, increase renal cGMP production under conditions of oxidative stress, thereby improving renal blood flow (RBF) and kidney function. EXPERIMENTAL APPROACH: Two novel sGC activators, runcaciguat and BAY-543, were tested on murine kidney. We measured cGMP levels in real time in kidney slices of cGMP sensor mice, vasodilation of pre-constricted glomerular arterioles and RBF in isolated perfused kidneys. Experiments were performed at baseline conditions, under L-NAME-induced NO deficiency, and in the presence of oxidative stress induced by ODQ. KEY RESULTS: Mouse glomeruli showed NO-induced cGMP increases. Under baseline conditions, sGC activator did not alter glomerular cGMP concentration or NO-induced cGMP generation. In the presence of ODQ, NO-induced glomerular cGMP signals were markedly reduced, whereas sGC activator induced strong cGMP increases. L-NAME and ODQ pretreated isolated glomerular arterioles were strongly dilated by sGC activator. sGC activator also increased cGMP and RBF in ODQ-perfused kidneys. CONCLUSION AND IMPLICATION: sGC activators increase glomerular cGMP, dilate glomerular arterioles and improve RBF under disease-relevant oxidative stress conditions. Therefore, sGC activators represent a promising class of drugs for chronic kidney disease treatment. LINKED ARTICLES: This article is part of a themed issue on cGMP Signalling in Cell Growth and Survival. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.11/issuetoc.


Subject(s)
Renal Insufficiency, Chronic , Vasodilation , Animals , Cyclic GMP , Female , Guanylate Cyclase , Humans , Kidney , Male , Mice , NG-Nitroarginine Methyl Ester , Nitric Oxide , Renal Insufficiency, Chronic/drug therapy , Soluble Guanylyl Cyclase
5.
Naunyn Schmiedebergs Arch Pharmacol ; 394(12): 2363-2379, 2021 12.
Article in English | MEDLINE | ID: mdl-34550407

ABSTRACT

Chronic kidney diseaQueryse (CKD) is associated with oxidative stress which can interrupt the nitric oxide (NO)/soluble guanylyl cyclase (sGC) signaling and decrease cyclic guanosine monophosphate (cGMP) production. Low cGMP concentrations can cause kidney damage and progression of CKD. The novel sGC activator runcaciguat targets the oxidized and heme-free form of sGC, restoring cGMP production under oxidative stress. The purpose of this study is to investigate if runcaciguat could provide an effective treatment for CKD. Runcaciguat was used for the treatment not only in rat CKD models with different etiologies and comorbidities, namely of hypertensive rats, the renin transgenic (RenTG) rat, and angiotensin-supplemented (ANG-SD) rat, but also in rats with diabetic and metabolic CKD, the Zucker diabetic fatty (ZDF) rat. The treatment duration was 2 to 42 weeks and runcaciguat was applied orally in doses from 1 to 10 mg/kg/bid. In these different rat CKD models, runcaciguat significantly reduced proteinuria (urinary protein to creatinine ratio; uPCR). These effects were also significant at doses which did not or only moderately decrease systemic blood pressure. Moreover, runcaciguat significantly decreased kidney injury biomarkers and attenuated morphological kidney damages. In RenTG rats, runcaciguat improved survival rates and markers of heart injury. These data demonstrate that the sGC activator runcaciguat exhibits cardio-renal protection at doses which did not reduce blood pressure and was effective in hypertensive as well as diabetic and metabolic CKD models. These data, therefore, suggest that runcaciguat, with its specific mode of action, represents an efficient treatment approach for CKD and associated CV diseases.


Subject(s)
Cyclopropanes , Diabetes Mellitus, Experimental , Hypertension , Renal Insufficiency, Chronic , Animals , Male , Rats , Blood Pressure/drug effects , Cyclic GMP/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activators/administration & dosage , Enzyme Activators/pharmacology , Hypertension/complications , Hypertension/drug therapy , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Zucker , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/prevention & control , Soluble Guanylyl Cyclase/drug effects , Soluble Guanylyl Cyclase/metabolism , Time Factors , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use
6.
J Med Chem ; 64(9): 5323-5344, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33872507

ABSTRACT

Herein we describe the discovery, mode of action, and preclinical characterization of the soluble guanylate cyclase (sGC) activator runcaciguat. The sGC enzyme, via the formation of cyclic guanosine monophoshphate, is a key regulator of body and tissue homeostasis. sGC activators with their unique mode of action are activating the oxidized and heme-free and therefore NO-unresponsive form of sGC, which is formed under oxidative stress. The first generation of sGC activators like cinaciguat or ataciguat exhibited limitations and were discontinued. We overcame limitations of first-generation sGC activators and identified a new chemical class via high-throughput screening. The investigation of the structure-activity relationship allowed to improve potency and multiple solubility, permeability, metabolism, and drug-drug interactions parameters. This program resulted in the discovery of the oral sGC activator runcaciguat (compound 45, BAY 1101042). Runcaciguat is currently investigated in clinical phase 2 studies for the treatment of patients with chronic kidney disease and nonproliferative diabetic retinopathy.


Subject(s)
Drug Design , Enzyme Activators/chemistry , Soluble Guanylyl Cyclase/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Dogs , Enzyme Activators/metabolism , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Half-Life , Heart Rate/drug effects , Hemodynamics/drug effects , Hypertension/drug therapy , Hypertension/pathology , Molecular Dynamics Simulation , Rats , Rats, Inbred SHR , Solubility , Soluble Guanylyl Cyclase/metabolism , Structure-Activity Relationship
7.
Int J Mol Sci ; 19(6)2018 Jun 09.
Article in English | MEDLINE | ID: mdl-29890734

ABSTRACT

Chronic Kidney Disease (CKD) is a highly prevalent disease with a substantial medical need for new and more efficacious treatments. The Nitric Oxide (NO), soluble guanylyl cyclase (sGC), cyclic guanosine monophosphate (cGMP) signaling cascade regulates various kidney functions. cGMP directly influences renal blood flow, renin secretion, glomerular function, and tubular exchange processes. Downregulation of NO/sGC/cGMP signaling results in severe kidney pathologies such as CKD. Therefore, treatment strategies aiming to maintain or increase cGMP might have beneficial effects for the treatment of progressive kidney diseases. Within this article, we review the NO/sGC/cGMP signaling cascade and its major pharmacological intervention sites. We specifically focus on the currently known effects of cGMP on kidney function parameters. Finally, we summarize the preclinical evidence for kidney protective effects of NO-donors, PDE inhibitors, sGC stimulators, and sGC activators.


Subject(s)
Kidney Diseases/pathology , Kidney/pathology , Nitric Oxide/metabolism , Signal Transduction , Soluble Guanylyl Cyclase/metabolism , Animals , Cyclic GMP/metabolism , Humans , Kidney Diseases/therapy
8.
Nephron ; 132(1): 59-69, 2016.
Article in English | MEDLINE | ID: mdl-26646377

ABSTRACT

BACKGROUND/AIMS: A critical involvement of the endocannabinoid/cannabinoid receptor system in diabetes and its complications has been recognized. Experimental evidence suggested that activation of the cannabinoid receptor type 2 (CB2), which is expressed in the kidney by podocytes and inflammatory cells, had a protective role in early streptozotocin-induced type 1 diabetes in mice. No experimental evidence is so far available on the effects of CB2 agonists in type 2 diabetes. In this study, we investigated the effects of a CB2 agonist given at a phase of overt disease on renal functional and structural changes in BTBR ob/ob mice, a model of type 2 diabetic nephropathy. METHODS: BTBR ob/ob mice received, from 10 to 21 weeks of age, vehicle, the selective CB2 agonist HU910, or lisinopril used as standard therapy for comparison. BTBR wild-type mice served as controls. RESULTS: Treatment with CB2 agonist reduced progressive albuminuria of BTBR ob/ob mice to a similar extent as ACE inhibitor. The antiproteinuric effect of CB2 agonist was associated with the amelioration of the defective nephrin expression in podocytes of diabetic mice. CB2 agonist limited mesangial matrix expansion, fibronectin accumulation and sclerosis. Glomerular infiltration of Mac-2-positive monocytes/machrophages was attenuated by CB2 agonist, at least in part due to the drug's ability to reduce MCP-1 chemotactic signals. Renoprotective effects of CB2 were similar to those achieved by ACE inhibitor. CONCLUSION: These results suggest that CB2 agonism is a potential option to be added to the available therapeutic armamentarium for type 2 diabetic nephropathy.


Subject(s)
Albuminuria/drug therapy , Albuminuria/etiology , Bridged Bicyclo Compounds/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/complications , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Receptor, Cannabinoid, CB2/agonists , Albuminuria/pathology , Animals , Blood Glucose/metabolism , Blood Pressure , Diabetic Neuropathies/pathology , Glomerular Filtration Rate , Kidney/pathology , Kidney Diseases/pathology , Male , Mice , Mice, Obese , Podocytes/pathology
9.
J Diabetes Res ; 2015: 404085, 2015.
Article in English | MEDLINE | ID: mdl-25785279

ABSTRACT

Diabetes mellitus is a lifelong, incapacitating metabolic disease associated with chronic macrovascular complications (coronary heart disease, stroke, and peripheral vascular disease) and microvascular disorders leading to damage of the kidneys (nephropathy) and eyes (retinopathy). Based on the current trends, the rising prevalence of diabetes worldwide will lead to increased cardiovascular morbidity and mortality. Therefore, novel means to prevent and treat these complications are needed. Under the auspices of the IMI (Innovative Medicines Initiative), the SUMMIT (SUrrogate markers for Micro- and Macrovascular hard end points for Innovative diabetes Tools) consortium is working on the development of novel animal models that better replicate vascular complications of diabetes and on the characterization of the available models. In the past years, with the high level of genomic information available and more advanced molecular tools, a very large number of models has been created. Selecting the right model for a specific study is not a trivial task and will have an impact on the study results and their interpretation. This review gathers information on the available experimental animal models of diabetic macrovascular complications and evaluates their pros and cons for research purposes as well as for drug development.


Subject(s)
Diabetes Complications/physiopathology , Diabetes Complications/therapy , Diabetes Mellitus, Experimental/therapy , Disease Models, Animal , Animals , Atherosclerosis/complications , Cardiovascular Diseases/complications , Cardiovascular Diseases/therapy , Clinical Trials as Topic , Coronary Artery Disease/complications , Diabetic Angiopathies/therapy , Humans , Hypoglycemic Agents/therapeutic use , Mice , Microcirculation , Models, Animal , Rats , Species Specificity
10.
J Bone Miner Metab ; 33(6): 625-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25534548

ABSTRACT

Pioglitazone, the peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonist is an effective therapy for type 2 diabetes, but has been associated with increased risk for bone fracture. Preclinical studies suggest that PPAR-α agonists (e.g., fenofibrate) increase bone mineral density/content, although clinical data on bone effects of fibrates are lacking. We investigated the effects of pioglitazone (10 mg/kg/day) and fenofibrate (25 mg/kg/day) on bone strength and bone histomorphometric parameters in osteopenic ovariectomized (OVX) rats. An additional group of rats received a combination of pioglitazone + fenofibrate to mimic the effects of a dual PPAR-α/γ agonist. The study consisted of a 13-week treatment phase followed by a 6-week treatment-free recovery period. Pioglitazone significantly reduced biomechanical strength at the lumbar spine and femoral neck compared with rats administered fenofibrate. Co-treatment with pioglitazone + fenofibrate had no significant effect on bone strength in comparison with OVX vehicle controls. Histomorphometric analysis of the proximal tibia revealed that pioglitazone suppressed bone formation and increased bone resorption at both cancellous and cortical bone sites relative to OVX vehicle controls. In contrast, fenofibrate did not affect bone resorption and only slightly suppressed bone formation. Discontinuation of pioglitazone treatment, both in the monotherapy and in the combination therapy arms, resulted in restoration of bone formation and resorption rates, demonstrating reversibility of effects. The above data support the concept that dual activation of PPAR-γ and PPAR-α attenuates the negative effects of PPAR-γ agonism on bone strength.


Subject(s)
Bone and Bones/pathology , Bone and Bones/physiopathology , Fenofibrate/administration & dosage , Fenofibrate/pharmacology , Ovariectomy , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology , Absorptiometry, Photon , Animals , Biomechanical Phenomena/drug effects , Compressive Strength/drug effects , Densitometry , Diaphyses/diagnostic imaging , Diaphyses/drug effects , Diaphyses/pathology , Diaphyses/physiopathology , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Femur/physiopathology , Femur Neck/diagnostic imaging , Femur Neck/drug effects , Femur Neck/pathology , Femur Neck/physiopathology , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Pioglitazone , Rats, Sprague-Dawley , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/pathology , Tibia/physiopathology , Tomography, X-Ray Computed
11.
J Med Chem ; 56(24): 9874-96, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24175572

ABSTRACT

We identified 6-alkoxy-5-aryl-3-pyridinecarboxamides as potent CB1 receptor antagonists with high selectivity over CB2 receptors. The series was optimized to reduce lipophilicity compared to rimonabant to achieve peripherally active molecules with minimal central effects. Several compounds that showed high plasma exposures in rats were evaluated in vivo to probe the contribution of central vs peripheral CB1 agonism to metabolic improvement. Both rimonabant and 14g, a potent brain penetrant CB1 receptor antagonist, significantly reduced the rate of body weight gain. However, 14h, a molecule with markedly reduced brain exposure, had no significant effect on body weight. PK studies confirmed similarly high exposure of both 14h and 14g in the periphery but 10-fold lower exposure in the brain for 14h. On the basis of these data, which are consistent with reported effects in tissue-specific CB1 receptor KO mice, we conclude that the metabolic benefits of CB1 receptor antagonists are primarily centrally mediated as originally believed.


Subject(s)
Amides/pharmacology , Pyridines/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Animals , CHO Cells , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Humans , Male , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
12.
J Endocrinol ; 212(2): 179-86, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22062085

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) γ agonists, such as pioglitazone (Pio), improve glycemia and lipid profile but are associated with bone loss and fracture risk. Data regarding bone effects of PPARα agonists (including fenofibrate (Feno)) are limited, although animal studies suggest that Feno may increase bone mass. This study investigated the effects of a 13-week oral combination treatment with Pio (10 mg/kg per day)+Feno (25 mg/kg per day) on body composition and bone mass parameters compared with Pio or Feno alone in adult ovariectomized (OVX) rats, with a 4-week bone depletion period, followed by a 6-week treatment-free period. Treatment of OVX rats with Pio+Feno resulted in ∼50% lower fat mass gain compared with Pio treatment alone. Combination treatment with Pio+Feno partially prevented Pio-induced loss of bone mineral content (∼45%) and bone mineral density (BMD; ∼60%) at the lumbar spine. Similar effects of treatments were observed at the femur, most notably at sites rich in trabecular bone. At the proximal tibial metaphysis, concomitant treatment with Pio+Feno prevented Pio exacerbation of ovariectomy-induced loss of trabecular bone, resulting in BMD values in the Pio+Feno group comparable to OVX controls. Discontinuation of Pio or Feno treatment of OVX rats was associated with partial reversal of effects on bone loss or bone mass gain, respectively, while values in the Pio+Feno group remained comparable to OVX controls. These data suggest that concurrent/dual agonism of PPARγ and PPARα may reduce the negative effects of PPARγ agonism on bone mass.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Fenofibrate/therapeutic use , Osteoporosis, Postmenopausal/drug therapy , PPAR alpha/agonists , PPAR gamma/agonists , Thiazolidinediones/adverse effects , Adiposity/drug effects , Animals , Biomarkers/blood , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Resorption/chemically induced , Bone Resorption/etiology , Bone and Bones/chemistry , Bone and Bones/drug effects , Collagen Type I/blood , Drug Therapy, Combination , Female , Fenofibrate/administration & dosage , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/therapeutic use , Osteocalcin/blood , Osteoporosis, Postmenopausal/blood , Osteoporosis, Postmenopausal/physiopathology , Ovariectomy , Peptides/blood , Pioglitazone , Random Allocation , Rats , Thiazolidinediones/administration & dosage , Thiazolidinediones/therapeutic use
13.
Bioorg Med Chem Lett ; 21(11): 3237-42, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21550236

ABSTRACT

A novel sulfonylureido pyridine series exemplified by compound 19 yielded potent inhibitors of FBPase showing significant glucose reduction and modest glycogen lowering in the acute db/db mouse model for Type-2 diabetes. Our inhibitors occupy the allosteric binding site and also extend into the dyad interface region of tetrameric FBPase.


Subject(s)
Aminopyridines/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fructose-Bisphosphatase/antagonists & inhibitors , Administration, Oral , Allosteric Site , Aminopyridines/chemistry , Animals , Crystallography, X-Ray , Diabetes Mellitus, Type 2 , Disease Models, Animal , Enzyme Inhibitors/chemistry , Fructose-Bisphosphatase/chemistry , Fructose-Bisphosphatase/metabolism , Humans , Inhibitory Concentration 50 , Liver/enzymology , Mice , Molecular Structure
14.
Cardiovasc Diabetol ; 10: 7, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21251281

ABSTRACT

BACKGROUND: Glycemic control and management of dyslipidemia to reduce cardiovascular risk are major therapeutic goals in individuals with type 2 diabetes mellitus (T2DM). This study was performed to evaluate the effects of aleglitazar, a balanced dual peroxisome proliferator-activated receptor α/γ (PPARα/γ) agonist, on both lipid and glycemic parameters in obese, hypertriglyceridemic, insulin-resistant rhesus monkeys. METHODS: A 135-day efficacy study was performed in six rhesus monkeys. After a 28-day baseline assessment (vehicle only), monkeys received oral aleglitazar 0.03 mg/kg per day for 42 days, followed by a 63-day washout period. Plasma levels of markers of glycemic and lipid regulation were measured at baseline, at the end of the dosing period, and at the end of the washout period. RESULTS: Compared with baseline values, aleglitazar 0.03 mg/kg per day reduced triglyceride levels by an average of 89% (328 to 36 mg/dL; P = 0.0035 when normalized for baseline levels) and increased high-density lipoprotein cholesterol levels by 125% (46 to 102 mg/dL; P = 0.0007). Furthermore, aleglitazar reduced low-density lipoprotein cholesterol levels (41%) and increased levels of apolipoprotein A-I (17%) and A-II (17%). Aleglitazar also improved insulin sensitivity by 60% (P = 0.001). Mean body weight was reduced by 5.9% from baseline values with aleglitazar at this dose (P = 0.043). CONCLUSIONS: Aleglitazar, a dual PPARα/γ agonist, has beneficial effects on both lipid and glucose parameters and may have a therapeutic role in modifying cardiovascular risk factors and improving glycemic control in patients with T2DM.


Subject(s)
Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Insulin Resistance , Metabolic Syndrome/drug therapy , Oxazoles/pharmacology , PPAR alpha/agonists , PPAR gamma/agonists , Prediabetic State/drug therapy , Thiophenes/pharmacology , Administration, Oral , Animals , Apolipoprotein A-I/blood , Apolipoprotein A-II/blood , Biomarkers/blood , Blood Glucose/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Disease Models, Animal , Eating , Glycated Hemoglobin/metabolism , Hypertriglyceridemia/blood , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/physiopathology , Hypoglycemic Agents/administration & dosage , Hypolipidemic Agents/administration & dosage , Macaca mulatta , Male , Metabolic Syndrome/blood , Metabolic Syndrome/physiopathology , Obesity/blood , Obesity/drug therapy , Obesity/physiopathology , Oxazoles/administration & dosage , PPAR alpha/metabolism , PPAR gamma/metabolism , Prediabetic State/blood , Prediabetic State/physiopathology , Thiophenes/administration & dosage , Time Factors , Triglycerides/blood , Weight Loss
15.
J Hypertens ; 28(8): 1676-86, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20498617

ABSTRACT

OBJECTIVE: The increased mortality observed with the cholesteryl ester transfer protein inhibitor torcetrapib is partly due to increased aldosterone production and blood pressure. The mechanisms underlying these effects were investigated. METHODS: Cytochrome P450 subunit 11B2 (aldosterone synthase), extracellular signal-regulated kinase (p44/42) and voltage-gated Cachannel alpha subunit mRNA profiling, aldosterone production, cytosolic calcium and RNA interference were assessed in adrenocarcinoma human cells (H295R). Telemetry was conducted in spontaneously hypertensive rats. RESULTS: Torcetrapib and angiotensin II (Ang II) but not dalcetrapib (a structurally different cholesteryl ester transfer protein inhibitor) elevated both cytochrome P450 subunit 11B2 mRNA and aldosterone production in H295R cells at 6 h. At days 1-5, torcetrapib produced a sustained increase of cytochrome P450 subunit 11B2 mRNA, unlike Ang II. Although torcetrapib and Ang II potentiated the effect of 25-OH cholesterol and raised pregnenolone levels, torcetrapib increased neither cytosolic Ca at 5 min nor extracellular signal-regulated kinase1/2 phosphorylation, suggesting initially divergent pathways. Unlike Ang II, torcetrapib steroidogenesis was not affected by Ang II type 1 receptor antagonism or voltage-gated T-type Ca channel antagonism, but was blocked by several L-type Cachannel antagonists. In unbiased genome-wide screening, Ang II and torcetrapib modulated an overlapping but distinct set of genes in H295R cells. Torcetrapib, but not Ang II, upregulated mRNA levels of the L-type Ca channel alpha 1C subunit. In spontaneously hypertensive rat, torcetrapib had a potent hypertensive effect mediated by the L-type Ca channel. CONCLUSION: The unique steroidogenic and hypertensive side effects of torcetrapib may be linked and involve voltage-gated L-type Ca channels. Structurally unrelated cholesteryl ester transfer protein inhibitors such as dalcetrapib do not share this effect.


Subject(s)
Anticholesteremic Agents/pharmacology , Calcium Channels, L-Type/drug effects , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Hypertension/drug therapy , Quinolines/pharmacology , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Adrenal Cortex/pathology , Adrenal Gland Neoplasms , Aldosterone/metabolism , Amides , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Cell Line, Tumor , Cytochrome P-450 CYP11B2/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Cytosol/drug effects , Cytosol/metabolism , Enzyme Induction/drug effects , Esters , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Humans , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Inbred SHR , Sodium Channels/metabolism , Structure-Activity Relationship , Sulfhydryl Compounds/pharmacology
17.
Bioorg Med Chem Lett ; 19(9): 2468-73, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19349176

ABSTRACT

Design, synthesis, and SAR of novel alpha-alkoxy-beta-arylpropionic acids as potent and balanced PPARalphagamma coagonists are described. One representative thereof, Aleglitazar ((S)-2Aa), was chosen for clinical development. Its X-ray structure in complex with both receptors as well as its high efficacy in animal models of T2D and dyslipidemia are also presented.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Oxazoles/chemical synthesis , Oxazoles/pharmacology , PPAR alpha/agonists , PPAR gamma/agonists , Thiophenes/chemical synthesis , Thiophenes/pharmacology , Animals , Chemistry, Pharmaceutical/methods , Crystallography, X-Ray/methods , Drug Design , Dyslipidemias/drug therapy , Humans , Inhibitory Concentration 50 , Ligands , Models, Chemical , Molecular Structure , Receptors, Cytoplasmic and Nuclear/metabolism
18.
ChemMedChem ; 4(6): 951-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19326383

ABSTRACT

An X-ray-guided design approach led to the identification of a novel, balanced class of alpha-ethoxy-phenylpropionic acid-derived dual PPARalpha/gamma agonists. The series shows a wide range of PPARalpha/gamma ratios within a rather narrow structural space. Advanced compounds possess favorable physicochemical and pharmacokinetic profiles and show a high efficacy in T2D and dyslipidemia animal models.


Subject(s)
Hypolipidemic Agents/chemistry , PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates/chemistry , Animals , Computer Simulation , Crystallography, X-Ray , Diabetes Mellitus, Type 2/drug therapy , Drug Design , Humans , Hypolipidemic Agents/chemical synthesis , Hypolipidemic Agents/pharmacokinetics , Mice , PPAR alpha/metabolism , PPAR gamma/metabolism , Phenylpropionates/chemical synthesis , Phenylpropionates/pharmacokinetics , Rats , Stereoisomerism , Structure-Activity Relationship
19.
J Med Chem ; 51(7): 2115-27, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-18335976

ABSTRACT

The application of the evolutionary fragment-based de novo design tool TOPology Assigning System (TOPAS), starting from a known CB1R (CB-1 receptor) ligand, followed by further refinement principles, including pharmacophore compliance, chemical tractability, and drug likeness, allowed the identification of benzodioxoles as a novel CB1R inverse agonist series. Extensive multidimensional optimization was rewarded by the identification of promising lead compounds, showing in vivo activity. These compounds reversed the CP-55940-induced hypothermia in Naval Medical Research Institute (NMRI) mice and reduced body-weight gain, as well as fat mass, in diet-induced obese Sprague-Dawley rats. Herein, we disclose the tools and strategies that were employed for rapid hit identification, synthesis and generation of structure-activity relationships, ultimately leading to the identification of (+)-[( R)-2-(2,4-dichloride-phenyl)-6-fluoro-2-(4-fluoro-phenyl)-benzo[1,3]dioxol-5-yl]-morpholin-4-yl-methanone ( R)-14g . Biochemical, pharmacokinetic, and pharmacodynamic characteristics of ( R)-14g are discussed.


Subject(s)
Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacology , Obesity/drug therapy , Receptor, Cannabinoid, CB1/agonists , Animals , Anti-Obesity Agents/chemistry , Benzodioxoles/chemical synthesis , Benzodioxoles/chemistry , Body Weight/drug effects , Crystallography, X-Ray , Cyclohexanols/antagonists & inhibitors , Cyclohexanols/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Humans , Hypothermia/chemically induced , Ligands , Male , Mice , Microsomes/drug effects , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/chemistry , Structure-Activity Relationship
20.
Obes Res ; 12(10): 1604-15, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15536224

ABSTRACT

OBJECTIVE: Magnetic resonance (MR) relaxometry has recently been introduced for noninvasive body composition analysis in awake mice. The purpose of the present study was to extend the method to rats and to introduce calibration procedures that render MR relaxometry fully quantitative. RESEARCH METHODS AND PROCEDURES: Proton T(2) MR relaxometry at 4.7 Tesla was used for body composition analyses in 700 awake mice and 400 rats of different strains and conditions. Relaxograms calculated from the signal decays observed with multi-spin-echo acquisition provided well-separated contributions of tissue water and fat. Analysis of fat composition was carried out in vivo using (13)C-MR spectroscopy. Evolution of body composition in rats was assessed during drug treatment. RESULTS: MR relaxometry for noninvasive body composition analysis in laboratory rodents was implemented on a standard MR scanner, and a throughput of >30 animals per hour was achieved. Excellent linearity and reproducibility with coefficients of variance as low as 2.5% and 1.7% were obtained in mice and rats, respectively. The lean mass-to-water ratio (mice, 1.35 +/- 0.03; rats, 1.39 +/- 0.04) and the proton density of fat (mice, 8.1 +/- 0.2; rats, 8.9 +/- 0.2 g/mol) were determined from cross-sectional data. Fat composition analysis by (13)C-MR spectroscopy corroborated these findings and yielded information on the average acyl chain length (16.3 +/- 1.6) and contributions of saturated (27 +/- 3%), monounsaturated (22 +/- 2%), and polyunsaturated (51 +/- 3%) fatty acids. Longitudinal assessments in rats treated with sibutramine and dexfenfluramine showed dose-related changes in body composition. DISCUSSION: T(2) MR relaxometry backed by solid calibration provides a powerful means for rapid quantitative body composition analysis in awake mice and rats that is suitable for serial investigations in pharmaceutical research.


Subject(s)
Anti-Obesity Agents/therapeutic use , Body Composition , Magnetic Resonance Spectroscopy/standards , Obesity/drug therapy , Animals , Calibration , Cross-Sectional Studies , Cyclobutanes , Dexfenfluramine , Magnetic Resonance Spectroscopy/methods , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Obese , Obesity/diet therapy , Rats , Rats, Sprague-Dawley , Rats, Wistar , Rats, Zucker , Reproducibility of Results , Sensitivity and Specificity , Weight Loss/physiology
SELECTION OF CITATIONS
SEARCH DETAIL