Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Article in English | MEDLINE | ID: mdl-37779395

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is an overwhelming neurodegenerative disease with progressive loss of memory. AD is characterized by the deposition of the senile plaques mainly composed of ß-amyloid (Aß) fragment, BDNF decline, Cholinergic system overactivity and neuroinflammation. Montelukast (MTK), a leukotriene receptor antagonist, showed astounding neuroprotective effects in a variety of neurodegenerative disorders. OBJECTIVE: This study aims to investigate the ameliorative effects of Montelukast in the scopolamineinduced Alzheimer's disease (AD) model in rats and evaluate its activity against neuroinflammation. METHODS: Thirty rats were split into five groups: Control group (1 mL/kg normal saline, i.p.), Montelukast perse (10 mg/kg, i.p.), Disease group treated with Scopolamine (3 mg/kg, i.p.), Donepezil group (3 mg/kg, i.p.), Montelukast treatment group (10 mg/kg, i.p.) and behavioural and biochemical tests were carried out to assess the neuro protective effect. RESULTS: Scopolamine treatment led to a significant reduction in learning and memory and an elevation in cholinesterase levels when compared with the control group (p < 0.01). Additionally, elevated oxidative stress and Amyloid-ß levels were associated with enhanced neuroinflammation (p < 0.05, p < 0.01). Furthermore, the decline in neurotrophic factor BDNF is also observed when compared with the normal control group (p < 0.01). Montelukast pre-treatment significantly attenuated learning and memory impairment and cholinesterase levels. Besides, Montelukast and standard drug donepezil administration significantly suppressed the oxidative stress markers (p < 0.01), Amyloid-ß levels, neuroinflammatory mediators (p < 0.05) and caused a significant increase in BDNF levels (p < 0.05) Conclusion: Montelukast bestowed ameliorative effects in scopolamine-induced AD animal models as per the previous studies via attenuation of memory impairment, cholinesterase neurotransmission, oxidative stress, Amyloid-ß levels, neuroinflammatory mediators and enhanced BDNF levels.

2.
ACS Chem Neurosci ; 14(18): 3444-3459, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37669120

ABSTRACT

Alzheimer's disease (AD) is one of the most prevalent and progressive neurodegenerative disorders, hallmarked by increased amyloid-ß deposition and enhanced oxidative load in the brain, ensuing cognitive decline. The present study is aimed at elucidating the neuroprotective effect of saroglitazar, a dual peroxisome-proliferator-activated receptor (PPARα/γ) agonist used in the treatment of diabetic dyslipidemia, against memory impairment induced by intraperitoneal scopolamine injection. 30 male Wistar rats were randomly divided into the following five groups: (A) Veh + Veh, (B) SGZ + Veh, (C) Veh + SCOP, (D) DPZ + SCOP, and (E) SGZ + SCOP. Rats of the respective groups were pretreated with saroglitazar (10 mg/kg, p.o.) and donepezil (3 mg/kg, p.o.) once daily for 16 days. During the final 9 days of the study, a daily injection of scopolamine (3 mg/kg, i.p.) was administered to the respective groups. Adjacent to the scopolamine injection, behavioral tests such as the open field, Y maze, novel object recognition test, and Morris water maze were conducted to assess learning and memory. Additionally, biochemical parameters such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), nitric oxide (NO), malondialdehyde (MDA), reduced glutathione (GSH), superoxide dismutase (SOD), brain-derived neurotrophic factor (BDNF), ß-amyloid levels, and NF-κB were measured in the hippocampus. The rats that received scopolamine injections showed significantly impaired short-term spatial and learning memory. This was associated with an increase in ß-amyloid, iNOS, nitric oxide (NO), malondialdehyde, NF-κB, and TNF-α levels in the hippocampus of AD rats. On the other hand, saroglitazar has provided promising data on its protective role in cognition by protecting the BDNF, SOD, and GSH decline. As a result, saroglitazar was found to be a promising therapy in AD by upregulating the antioxidant status and cholinergic activity and preventing memory loss. Collectively, findings in the present study revealed that saroglitazar protected AD by suppressing scopolamine-mediated learning and memory deficits, oxidative stress, and cholinergic damage. Studying these mechanisms may conclude the protective role of saroglitazar against AD. However, further studies in transgenic animals will provide numerous insights into treatment mechanisms and contribute to developing a therapeutic intervention for AD.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Male , Animals , Rats , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Scopolamine , Brain-Derived Neurotrophic Factor , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , NF-kappa B , Acetylcholinesterase , Butyrylcholinesterase , Nitric Oxide , Rats, Wistar , Amyloid beta-Peptides
3.
Eur J Pharmacol ; 934: 175320, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36220360

ABSTRACT

Ever Since, pharmaceutical companies are facing challenges to develop new drug products faster and economical with good quality, safety and efficacy. The advent of Artificial intelligence (AI) with computational technology empowers scientists, impacts society at a great scale by developing new drugs at rapid pace. Artificial intelligence is the science and engineering of creating intelligent machines using personified knowledge. There are many opportunities to apply AI tools to the drug discovery pipeline. Examples include target identification, identification of biomarkers, molecular modelling, synthesis of molecules, predicting toxicity and picking up leads. Further, this technology also helps the clinical scientists in clinical trial design, execution and real-time analysis. Altogether it facilitates the process of drug discovery, development and also provides better therapy to the patients. Apart from drug discovery and development, AI also has applications in the area of diagnosis, drug delivery, patient adherence and better monitoring of safety. There are many instances where AI can perform tasks better than humans and aid healthcare providers in treating patients. In this article, we have provided discussion on how AI is advancing the health care field to achieve greater success.


Subject(s)
Artificial Intelligence , Delivery of Health Care , Humans , Drug Discovery , Pharmaceutical Preparations
4.
Eur J Pharmacol ; 927: 175054, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35636524

ABSTRACT

PI3Kδ plays a critical role in adaptive immune cell activation and function. Suppression of PI3Kδ has been shown to counter excessive triggering of immune responses which has led to delineating the role of this isoform in the pathophysiology of autoimmune disorders. In the current study, we have described preclinical characterization of PI3Kδ specific inhibitor LL-00071210 in various rheumatoid arthritis models. LL-00071210 displayed excellent in vitro potency in biochemical and cellular assay against PI3Kδ with IC50 values of 24.6 nM and 9.4 nM, respectively. LL-00071210 showed higher selectivity over PI3Kγ and PI3Kß as compared to available PI3K inhibitors. LL-00071210 had good stability in liver microsomes and plasma across species and showed low clearance, low-to-moderate Vss, with bioavailability of >50% in preclinical species. LL-00071210 demonstrated excellent in vivo efficacy in adjuvant-induced and collagen-induced arthritis models. Co-administration of LL-00071210 and methotrexate at subtherapeutic dose regimen in collagen induced arthritis model led to additive effects, indicating the combination potential of LL-00071210 along with available disease modifying anti-rheumatic drugs (DMARD). In conclusion, we have described a specific PI3Kδ inhibitor with ∼100-fold selectivity over other PI3K isoforms. LL-00071210 has good drug-like properties and thus warrants testing in the clinic for the treatment of autoimmune diseases.


Subject(s)
Arthritis, Rheumatoid , Phosphatidylinositol 3-Kinases , Arthritis, Rheumatoid/drug therapy , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Structure-Activity Relationship
5.
J Med Chem ; 64(23): 17004-17030, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34843241

ABSTRACT

The role of calcium release-activated calcium (CRAC) channels is well characterized and is of particular importance in T-cell function. CRAC channels are involved in the pathogenesis of several autoimmune diseases, making it an attractive therapeutic target for treating inflammatory diseases, like rheumatoid arthritis (RA). A systematic structure-activity relationship study with the goal of optimizing lipophilicity successfully yielded two lead compounds, 36 and 37. Both compounds showed decent potency and selectivity and a remarkable pharmacokinetic profile. Further characterization in in vivo RA models and subsequent histopathological evaluation of tissues led to the identification of 36 as a clinical candidate. Compound 36 displayed an excellent safety profile and had a sufficient safety margin to qualify it for use in human testing. Oral administration of 36 in Phase 1 clinical study in healthy volunteers established favorable safety, tolerability, and good target engagement as measured by levels of IL-2 and TNF-α.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Release Activated Calcium Channels/antagonists & inhibitors , Calcium/metabolism , Drug Discovery , Administration, Oral , Animals , Area Under Curve , Arthritis, Rheumatoid/drug therapy , Calcium Channel Blockers/pharmacokinetics , Clinical Trials, Phase I as Topic , Humans , Jurkat Cells , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Inbred Lew , Structure-Activity Relationship
6.
Xenobiotica ; 49(5): 528-539, 2019 May.
Article in English | MEDLINE | ID: mdl-29771166

ABSTRACT

The present study was aimed at developing simplified physiologically based semi-mechanistic algorithms to predict Vss and interspecies scaling factors to predict tissue-Kps which require minimum input parameters, diminish the computing complexity and have better predictability. Vss of 86 structurally diverse compounds in preclinical species and 27 compounds in humans were predicted using only lung- and muscle-Kp as inputs. Interspecies scaling factor (s) were developed based on fold-differences in individual tissue lipid contents, relative organ blood flow: relative organ weight ratio between two species. Tissue-Kps were predicted for 34 compounds using the newly developed interspecies scaling factors. The predicted-to-experimental Vss values for all the 113 compounds was 1.3 ± 0.9 with 83% values being within a factor of two. The tissue-Kps in rat, dog and human were predicted using experimental tissue-Kp data in rodents and interspecies scaling factors and here also, 83% of tissue-Kps were within two-fold of the experimental values. In conclusion, simplified physiologically based algorithms have been developed to predict both volume of distribution and tissue-Kps, in which required input parameters as well as computing complexity have been noticeably reduced.


Subject(s)
Algorithms , Computer Simulation , Lung/metabolism , Models, Biological , Muscle, Skeletal/metabolism , Animals , Humans , Organ Size
7.
Med Mol Morphol ; 52(1): 36-43, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29959534

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is characterized by the presence of hepatic steatosis, oxidative stress, inflammation, and hepatocyte injury with or without fibrosis. In this study, we explored the effect of APD668, a GPR119 agonist alone or in combination with linagliptin, a DPPIV inhibitor, on the progression of steatohepatitis in a murine model of NASH with diabetes. A novel NASH model with diabetes was generated by administration of streptozotocin injection to neonatal C57BL/6 mice (2-3 days old) combined with a high-fat diet feeding from the age of 4 weeks. The plasma biochemical parameters, oxidative stress, inflammation and histopathological changes were assessed. APD668 alone showed reduction in plasma glucose (- 39%, P < 0.05) and triglyceride level (- 26%) whereas a combined treatment of APD668 with linagliptin resulted in a more pronounced reduction in plasma glucose (- 52%, P < 0.001) and triglyceride (- 50%, P < 0.05) in NASH mice. In addition, co-administration of APD668 with linagliptin demonstrated a significant decrease in hepatic triglyceride, NAS score, hepatic TBARS and hepatic TNF-α in NASH mice with diabetes. These findings suggest that GPR119 receptor agonists in combination with DPPIV inhibitors may represent a promising therapeutic strategy for the treatment of NASH.


Subject(s)
Diabetes Mellitus, Experimental/complications , Linagliptin/pharmacology , Non-alcoholic Fatty Liver Disease/prevention & control , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Diet, High-Fat , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Progression , Fibrosis , Inflammation , Linagliptin/therapeutic use , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Receptors, G-Protein-Coupled/agonists
8.
Expert Opin Ther Targets ; 22(7): 615-628, 2018 07.
Article in English | MEDLINE | ID: mdl-29911915

ABSTRACT

INTRODUCTION: Incretin hormones, glucose-dependent insulinotropic polypeptide (GIP), and glucagon-like peptide-1 (GLP-1) exert pleiotropic effects on endocrine pancreas and nervous system. Expression of GIP and GIP receptor (GIPR) in neurons, their roles in neurogenesis, synaptic plasticity, neurotransmission, and neuromodulation uniquely position GIPR for therapeutic applications in neurodegenerative disorders. GIP analogs acting as GIPR agonists attenuate neurobehavioral and neuropathological sequelae of neurodegenerative disorders in preclinical models, e.g. Alzheimer's disease (AD), Parkinson's disease (PD), and cerebrovascular disorders. Modulation of GIPR signaling offers an unprecedented approach for disease modification by arresting neuronal viability decline, enabling neuronal regeneration, and reducing neuroinflammation. Growth-promoting effects of GIP signaling and broad-based neuroprotection highlight the therapeutic potential of GIPR agonists. Areas covered: This review focuses on the role of GIPR-mediated signaling in the central nervous system in neurophysiological and neuropathological conditions. In context of neurodegeneration, the article summarizes potential of targeting GIPR signaling for neurodegenerative conditions such as AD, PD, traumatic brain injury, and cerebrovascular disorders. Expert opinion: GIPR represents a validated therapeutic target for neurodegenerative disorders. GIPR agonists impart symptomatic improvements, slowed neurodegeneration, and enhanced neuronal regenerative capacity in preclinical models. Modulation of GIPR signaling is potentially a viable therapeutic approach for disease modification in neurodegenerative disorders.


Subject(s)
Gastric Inhibitory Polypeptide/analogs & derivatives , Neurodegenerative Diseases/physiopathology , Receptors, Gastrointestinal Hormone/agonists , Animals , Drug Development/methods , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Incretins/metabolism , Neurodegenerative Diseases/drug therapy , Receptors, Gastrointestinal Hormone/metabolism , Signal Transduction/drug effects
9.
Eur J Pharmacol ; 828: 31-41, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29577894

ABSTRACT

Huntington's disease (HD) is an inherited complex progressive neurodegenerative disorder with an established etiopathology linked to neuronal oxidative stress and corticostriatal excitotoxicity. Present study explores the effects of glucose-dependent insulinotropic polypeptide (GIP) receptor agonist on the neurobehavioral sequelae of quinolinic acid-induced phenotype of Huntington's disease in rats. Bilateral administration of quinolinic acid (300 nmol/4 µl) to the rat striatum led to characteristic deficits in, locomotor activity, motor coordination, neuromuscular coordination and short-term episodic memory. Therapeutic treatment for 14 days with a stable and brain penetrating GIP receptor agonist, D-Ala2GIP (100 nmol/kg, i.p.), attenuated the neurobehavioral deficits due to quinolinic acid (QA) administration. Protective actions of D-Ala2GIP were sensitive to blockade with a GIP receptor antagonist, (Pro3)GIP (50 nmol/kg, i.p.), indicating specific involvement of GIP receptor signaling pathway. Stimulation of GIP receptor with D-Ala2GIP attenuated lipid peroxidation, evidenced by reduced levels of brain malondialdehyde (MDA), and restoration of reduced glutathione (GSH) levels in brain. Quinolinic acid administration led to significant loss of striatal monoamines, e.g., norepinephrine, epinephrine, serotonin, dopamine, and metabolites, 3,4-Dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA) and 5-Hydroxyindoleacetic acid (5-HIAA). D-Ala2GIP attenuated the QA-induced depletion of striatal monoamines, without affecting the monoamine degradation pathways. Thus, observed effects with D-Ala2GIP in the QA-induced Huntington's disease model could be attributable to reduction in lipid peroxidation, restoration of endogenous antioxidants and decreased striatal monoamine levels. These findings together suggest that stimulation of GIP receptor signaling pathway in brain could be a potential therapeutic strategy in the symptomatic management of Huntington's disease.


Subject(s)
Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Huntington Disease/metabolism , Lipid Peroxidation/drug effects , Neostriatum/metabolism , Receptors, Gastrointestinal Hormone/agonists , Animals , Body Weight/drug effects , Disease Models, Animal , Drug Interactions , Gait/drug effects , Gastric Inhibitory Polypeptide/chemistry , Glutathione/metabolism , Hand Strength , Huntington Disease/chemically induced , Huntington Disease/physiopathology , Locomotion/drug effects , Male , Malondialdehyde/metabolism , Memory, Episodic , Neostriatum/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Quinolinic Acid/adverse effects , Rats , Rats, Wistar
10.
Biochem Biophys Res Commun ; 495(2): 1608-1613, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29203247

ABSTRACT

Non-Alcoholic SteatoHepatitis (NASH) is the more severe form of Non-Alcoholic Fatty Liver Disease (NAFLD) and is characterized by the presence of hepatic steatosis, oxidative stress, inflammation, hepatocyte injury with or without fibrosis. Recently, GPR119 receptor has emerged as a novel therapeutic target for the treatment of dyslipidemia and non-alcoholic steatohepatitis. In the present study, we investigated the effect of APD668, a GPR119 agonist alone or in combination with linagliptin, a DPPIV inhibitor on the progression of steatohepatitis in mice fed on a high trans-fat diet. In this study, monotherapy with either APD668 or linagliptin caused a reduction in the levels of ALT, AST, glucose, cholesterol and epididymal fat mass but the effect was more pronounced upon treatment with combination of both drugs. On the other hand, combined treatment of APD668 with linagliptin demonstrated a non-significant additive effect in reduction of hepatic triglyceride (-78%) and cholesterol (-56%) compared to monotherapy groups. Moreover, co-administration of APD668 and linagliptin resulted in enhanced levels of active GLP-1 with additional benefit of significant synergistic decrease in body weight gain (-19%) in mice. We speculated that the enhanced effect observed with the combination treatment could be due to either 1) direct activation of GPR119 receptors present in liver and intestine or 2) enhanced active GLP-1 levels or 3) decreased degradation of GLP-1 in-vivo through DPPIV inhibition. Therefore, these findings clearly suggest that GPR119 receptor agonists in combination with DPPIV inhibitors may represent a promising therapeutic strategy for the treatment of non-alcoholic steatohepatitis.


Subject(s)
Linagliptin/administration & dosage , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Animals , Diet, High-Fat/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Disease Models, Animal , Disease Progression , Drug Synergism , Drug Therapy, Combination , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, G-Protein-Coupled/agonists , Weight Gain/drug effects
11.
Eur J Drug Metab Pharmacokinet ; 43(3): 331-346, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29250739

ABSTRACT

BACKGROUND: Predicting target site drug concentrations is of key importance for rank ordering compounds before proceeding to chronic pharmacodynamic models. We propose generic tumor-specific correlation-based regression equations to predict tumor-to-plasma ratios (tumor-Kps) in slow- and fast-growing xenograft mouse models. METHODS: Disposition of 14 basic small molecules was investigated extensively in mouse plasma, tissues and tumors after a single oral dose administration. Linear correlation was assessed and compared between tumor-Kp and normal tissue-to-plasma ratio (tissue-Kps) separately for each tumor xenograft. The developed regression equations were validated by leave-one-out cross-validation (LOOCV) method. RESULT: Both slow- and fast-growing tumor-Kps showed good correlation (r 2 ≥ 0.7) with majority of the normal tissue-Kps. Substantial difference was observed in the slopes of developed equations between two xenografts, which was in line with observed difference in tumor distribution. The linear correlations between tumor-Kp and skin- or spleen-Kp were within the acceptable statistical criteria (LOOCV) across xenografts and the class of compounds evaluated. Since > 70% of tumor-Kps from the test data sets were predicted within a factor of twofold for both slow- and fast-growing xenograft mouse models, the results validate the applicability of the developed equations across xenografts. CONCLUSION: Tumor-specific correlation-based regression equations were developed and their applicability was adequately validated across xenografts. These equations could be successfully translated to predict tumor concentrations in order to preclude experimental tumor-Kp determination.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Plasma/metabolism , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacokinetics , Animals , Disease Models, Animal , Female , Heterografts , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/blood , Neoplasms, Experimental/blood , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Tissue Distribution
12.
Eur J Pharmacol ; 804: 38-45, 2017 Jun 05.
Article in English | MEDLINE | ID: mdl-28366809

ABSTRACT

The aim of the present study was to evaluate the ability of D-Ala2GIP, a gastric inhibitory polypeptide (GIP) receptor agonist, to attenuate the behavioral phenotype of Parkinson's disease caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in mice. In the behavioral studies, MPTP administration led to spontaneous locomotor activity deficits, impaired rotarod performance, akinesia, muscular rigidity and increased tremor amplitude, which was attenuated by pretreatment with D-Ala2GIP (50-100 nmol/kg, i.p.). This acute neuroprotective response by D-Ala2GIP was found to be blocked by a selective GIP receptor antagonist, (Pro3)GIP (50 nmol/kg, i.p.), indicating that the observed effects are mediated through GIP receptor mediated signaling pathway. Biochemical studies revealed that D-Ala2GIP reduced the brain malondialdehyde levels and enhanced the brain glutathione levels, thereby mitigating the MPTP-induced oxidative stress. MPTP administration resulted in reduction of the striatal concentration of dopamine and its metabolites, homovanillic acid (HVA) and 3, 4-Dihydroxyphenylacetic acid (DOPAC). Pretreatment with D-Ala2GIP attenuated the loss of striatal dopamine levels without affecting the normal dopamine catabolism. Thus, the observed effects in the MPTP-induced Parkinsonism model could be in part attributable to the antioxidant properties of D-Ala2GIP and enhanced turnover of dopamine in the nigrostriatal pathways in mouse brain. These findings together suggest that GIP receptor could be a therapeutic target in the management of symptoms of Parkinson's disease.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Gastric Inhibitory Polypeptide/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Gastrointestinal Hormone/agonists , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Dopamine/metabolism , Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/therapeutic use , Glutathione/metabolism , Homovanillic Acid/metabolism , Locomotion/drug effects , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/pathology , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Tremor/drug therapy
13.
Eur J Pharmacol ; 801: 35-45, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28274625

ABSTRACT

G-protein coupled receptor 119 (GPR119) receptor is a rhodopsin-like, class A Gαs-coupled receptor, predominantly expressed in pancreatic islet cells and intestinal entero-endocrine cells. GPR119 has been emerged as a novel therapeutic target for the treatment of dyslipidemia in type 2 diabetes. In this study, we investigated the effect of APD668, a GPR119 agonist alone and in combination with linagliptin, a DPPIV inhibitor on oral fat tolerance test. Our findings demonstrate that APD668, a GPR119 agonist inhibits the intestinal triglyceride absorption after acute fat load in mice. Single dose administration of APD668 increases incretin secretion and enhances total PYY levels in presence of fat load in mice. We found that, the anti-dyslipidemic action of APD668 was reversed in presence of exendin-3 in oral fat tolerance test. In addition, our results showed that exendin-3 (9-39) failed to block the effect of APD668 on gastric emptying indicating that gastric emptying effects of APD668 are indeed mediated through GPR119 receptor dependent mechanism. Combined administration of APD668 and linagliptin significantly increased plasma active GLP-1 levels in-vivo and showed improvement in fat tolerance. However, APD668 failed to show anti-dyslipidemic activity in tyloxapol-induced hyperlipidemia in mice. Furthermore, we investigated the chronic effects of APD668 on hepatic steatosis in high trans-fat diet fed steatohepatitis model in mice. Oral administration of APD668 in HTF diet fed mice ameliorated hepatic endpoints such as plasma ALT, AST, liver weight and steatosis. These findings suggest that GPR119 agonists may represent a promising therapeutic strategy for the treatment of dyslipidemia and non-alcoholic steatohepatitis.


Subject(s)
Diet, High-Fat/adverse effects , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Drug Interactions , Gastric Emptying/drug effects , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Linagliptin/pharmacology , Male , Mesylates/pharmacology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Oxadiazoles/pharmacology , Polyethylene Glycols/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Tetrazoles/pharmacology , Thiazoles/pharmacology , Triglycerides/blood
14.
Eur J Drug Metab Pharmacokinet ; 42(5): 835-847, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28194579

ABSTRACT

BACKGROUND: Majority of reported studies so far developed correlation regression equations using the rat muscle-to-plasma drug concentration ratio (Kp-muscle) to predict tissue-to-plasma drug concentration ratios (Kp-tissues). Use of regression equations derived from rat Kp-muscle may not be ideal to predict the mice tissue-Kps as there are species differences. OBJECTIVES: (i) To develop the linear regression equations using mouse tissue-Kps; (ii) to assess the correlation between organ blood flow and/or organ weight with tissue-Kps and (iii) compare the observed tissue-Kps from mice with corresponding predicted tissue-Kps using Richter's rat-Kp specific equations. METHOD: Disposition of 12 small molecules were investigated extensively in mouse plasma and tissues after a single oral dose administration. Linear correlation was assessed for each of the tissue with rest of the other tissues, separately for weak and strong bases. RESULT: Newly developed regression equations using mice tissue-Kps, predicted 79% data points within twofold. As observed correlation r 2 range was 0.75-0.98 between Kp-muscle and Kp-brain, -spleen, -skin, -liver, -lung, suggesting superior correlation between the tissue-Kps. Order of tissue-Kps, showed that tissue concentrations were directly proportional to the organ blood flow and inversely to the organ weight. Further, the observed tissue-Kps from mice were compared with corresponding predicted tissue-Kps using Richter's rat-Kp specific equations. Overall, 46, 54 and 63% data points were under predicted (<0.5-fold) for liver, spleen and lung, respectively. Whereas 63 and 75% data points were over predicted (>twofold) for skin and brain, respectively. These findings suggest that cross species extrapolation predictability is poor. CONCLUSION: All these findings together suggest that mouse specific regression equations developed under controlled experimental conditions could be most appropriate for predicting mouse tissue-Kps for compounds with wide range of volume of distribution.


Subject(s)
Pharmaceutical Preparations/metabolism , Plasma/metabolism , Animals , Brain/metabolism , Female , Linear Models , Liver/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , Muscles/metabolism , Rats , Tissue Distribution
15.
Bioorg Med Chem Lett ; 24(24): 5587-5592, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25466180

ABSTRACT

Nitric oxide-releasing non-steroidal anti-inflammatory drugs (NO-NSAIDs) are gaining attention as potentially gastric-sparing NSAIDs. Herein, we report a novel class of '1-(nitrooxy)ethyl ester' group-containing NSAIDS as efficient NO releasing 'true' prodrugs of aspirin and naproxen. While an aspirin prodrug exhibited comparable oral bioavailability and antiplatelet activity (i.e., TXB2 inhibition) to those of aspirin, a naproxen prodrug exhibited better bioavailability than naproxen. These promising NO-NSAIDs protected experimental rats from gastric damage. We therefore believe that these promising NO-NSAIDs could represent a new class of potentially 'Safe NSAIDs' for the treatment of arthritic pain, inflammation and cardiovascular disorders in the case of NO-aspirin.


Subject(s)
Aspirin/analogs & derivatives , Naproxen/analogs & derivatives , Nitrates/chemistry , Prodrugs/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Area Under Curve , Aspirin/chemistry , Aspirin/pharmacokinetics , Aspirin/pharmacology , Aspirin/toxicity , Blood Platelets/drug effects , Blood Platelets/enzymology , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Drug Design , Drug Stability , Gastric Mucosa/drug effects , Half-Life , Humans , Naproxen/chemistry , Naproxen/pharmacokinetics , Naproxen/pharmacology , Naproxen/toxicity , Nitrates/pharmacokinetics , Nitrates/pharmacology , Nitrates/toxicity , Nitric Oxide/metabolism , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/toxicity , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/toxicity , ROC Curve , Rats , Rats, Sprague-Dawley , Thromboxane B2/metabolism
16.
Eur J Med Chem ; 65: 337-47, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23735282

ABSTRACT

Biphenyl carboxylic acids, exemplified by compound 5, are known potent inhibitors of diacylglycerol acyltransferase, DGAT1, an enzyme involved in the final committed step of triglyceride biosynthesis. We have synthesized and evaluated 2-phenylthiazole, 4-phenylthiazole, and 5-phenylthiazole analogs as DGAT1 inhibitors. The 5-phenylthiazole series exhibited potent DGAT1 inhibition when evaluated using an in vitro enzymatic assay and an in vivo fat tolerance test in mice. Compound 33 (IC50 = 23 nM) exhibiting promising oral pharmacokinetic parameters (AUCinf = 7058 ng h/ml, T1/2 = 0.83 h) coupled with 87 percent reduction of plasma triglycerides in vivo may serve as a lead for developing newer anti-obesity agents.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Thiazoles/pharmacology , Triglycerides/antagonists & inhibitors , Administration, Oral , Animals , Diacylglycerol O-Acyltransferase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Mice , Molecular Structure , Structure-Activity Relationship , Thiazoles/administration & dosage , Thiazoles/chemistry , Triglycerides/blood
17.
Bioorg Med Chem Lett ; 23(3): 834-8, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265878

ABSTRACT

We report our attempts at improving the oral efficacy of low-nanomolar inhibitors of xanthine oxidase from isocytosine series through chemical modifications. Our lead compound had earlier shown good in vivo efficacy when administered intraperitoneally but not orally. Several modifications are reported here which achieved more than twofold improvement in exposure. A compound with significant improvement in oral efficacy was also obtained.


Subject(s)
Cytosine/analogs & derivatives , Enzyme Inhibitors/chemistry , Xanthine Oxidase/antagonists & inhibitors , Administration, Oral , Animals , Catalytic Domain , Cytosine/administration & dosage , Cytosine/chemistry , Cytosine/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Models, Animal , Models, Molecular , Molecular Structure , Rats
18.
Bioorg Med Chem Lett ; 22(24): 7543-6, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23122864

ABSTRACT

Structure-activity relationship studies were carried out for lead generation following structure-guided design approach from an isocytosine scaffold identified earlier for xanthine oxidase inhibition. A 470-fold improvement in in vitro IC(50) was obtained in the process. Five most potent compounds with nanomolar IC(50) values were selected for pharmacokinetics and in vivo experiments. The best compound showed good in vivo activity when administered intraperitoneally but was not active by oral route. The results suggest that improvement in oral exposure could improve the in vivo efficacy of this series.


Subject(s)
Cytosine/analogs & derivatives , Disease Models, Animal , Drug Design , Enzyme Inhibitors/pharmacology , Hyperuricemia/drug therapy , Xanthine Oxidase/antagonists & inhibitors , Administration, Oral , Animals , Cytosine/administration & dosage , Cytosine/chemical synthesis , Cytosine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Hyperuricemia/enzymology , Hyperuricemia/metabolism , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship , Time Factors , Xanthine Oxidase/metabolism
19.
Eur J Med Chem ; 54: 324-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22683241

ABSTRACT

Diacylglycerol acyltransferase, DGAT1, is a promising target enzyme for obesity due to its involvement in the committed step of triglyceride biosynthesis. Amino biphenyl carboxylic acids, exemplified by compound 4, are known potent inhibitors of hDGAT1. However the high cLogP and poor solubility of these biphenyl analogs might tend to limit their development. We have synthesized and evaluated compounds containing 3-phenylisoxazole, 5-phenyloxazole, and 3-phenyl-1,2,4-oxadiazole biaryl units for their hDGAT1 inhibition. Our aim in synthesizing such heterocyclic analogs was to improve the cLogP and solubility of these molecules while retaining hDGAT1 potency. Several compounds within the 3-phenylisoxazole series exhibited potent hDGAT1 inhibition when evaluated using an in vitro enzymatic assay. Certain promising compounds were studied for their potential to reduce triglyceride levels using an in vivo fat tolerance test in mice and were also evaluated for any possible improvement to their solubility. Compound 40a (IC(50) = 64 nM) with an in vivo plasma triglyceride reduction of 90 percent, and a solubility of 0.43 mg/ml at pH 7.4 may serve as a new lead for developing newer anti-obesity agents.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Isoxazoles/chemical synthesis , Isoxazoles/pharmacology , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Urea/chemistry , Animals , Chemistry Techniques, Synthetic , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Isoxazoles/chemistry , Mice , Oxadiazoles/chemistry , Solubility , Structure-Activity Relationship , Water/chemistry
20.
Bioorg Med Chem ; 20(9): 2930-9, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22483591

ABSTRACT

In recent years, xanthine oxidase has emerged as an important target not only for gout but also for cardiovascular and metabolic disorders involving hyperuricemia. Contrary to popular belief, recent clinical trials with uricosurics have demonstrated that enhanced excretion of uric acid is, by itself, not adequate to treat hyperuricemia; simultaneous inhibition of production of uric acid by inhibition of xanthine oxidase is also important. Virtual screening of in-house synthetic library followed by in vitro and in vivo testing led to the identification of a novel scaffold for xanthine oxidase inhibition. In vitro activity results corroborated the results from molecular docking studies of the virtual screening hits. The isocytosine scaffold maintains key hydrogen bonding and pi-stacking interactions in the deep end of the xanthine-binding pocket, which anchors it in an appropriate pose to inhibit binding of xanthine and shows promise for further lead optimization using structure-based drug design approach.


Subject(s)
Computer Simulation , Cytosine/analogs & derivatives , Enzyme Inhibitors/chemistry , Xanthine Oxidase/antagonists & inhibitors , Animals , Cytosine/chemical synthesis , Cytosine/chemistry , Cytosine/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Hyperuricemia/chemically induced , Hyperuricemia/drug therapy , Male , Oxonic Acid/pharmacology , Oxonic Acid/toxicity , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Xanthine Oxidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...