Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Peptides ; 159: 170902, 2023 01.
Article in English | MEDLINE | ID: mdl-36375661

ABSTRACT

Pancreastatin (PST) is an endogenous bioactive peptide. PST is generated from chromogranin A (Chga) protein which is released by chromaffin and neuroendocrine cells. PST exhibits diabetogenic effect by antagonizing the action of insulin in adipocytes. The level of PST rises during obesity, resulting in persistent low-grade inflammation in adipocytes. Pancreastatin inhibitor 8 (PSTi8), which is developed by modification of PST sequence which antagonizes the action of PST. In this study, we investigated the immunometabolic effect of PSTi8 in the diet-induced obesity (DIO) model in C57BL/6 mice. Here we found PSTi8 decreased the body weight gain, fat mass and increased the lean mass in (DIO) mice. It also showed reduction of adipocyte hypertrophy in eWAT and lipid accumulation in liver of DIO mice. Immunoprofiling of stromal vascular fraction isolated from eWAT of PTSi8 treated mice showed increased anti-inflammatory M2 macrophages, Eosinophil, T-regulatory cells and reduced pro-inflammatory M1 macrophages, CD4 and CD8 T cell population. Apart from this, PSTi8 also improved the mitochondrial function by decreasing reactive oxygen species and increasing mitochondrial membrane potential, NADPH/NADP ratio and citrate synthase activity in eWAT of DIO mice. It also increased the protein expression of pAMPK, pAKT, Arginase -1 and decreased the expression of MHC-II and iNOS in eWAT of DIO mice. In conclusion, PSTi8 exerted its beneficial effect on restoring energy expenditure by reducing adipose tissue inflammation.


Subject(s)
Diet, High-Fat , Insulin Resistance , Mice , Animals , Chromogranin A , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Adipose Tissue/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Obesity/drug therapy , Obesity/metabolism , Homeostasis
2.
Pharmacology ; 107(1-2): 54-68, 2022.
Article in English | MEDLINE | ID: mdl-34814141

ABSTRACT

INTRODUCTION: The present study deals with the synthesis of pregnane-oximino-amino-alkyl-ethers and their evaluation for antidiabetic and anti-dyslipidemic activities in validated animal and cell culture models. METHODS: The effect on glucose tolerance was measured in sucrose-loaded rats; antidiabetic activity was evaluated in streptozotocin (STZ)-induced diabetic rats and genetically diabetic db/db mice; the anti-dyslipidemic effect was characterized in high-fructose, high-fat diet (HFD)-fed dyslipidemic hamsters. The effect on glucose production and glucose utilization was analyzed in HepG2 liver and L6 skeletal muscle cells, respectively. RESULTS: From the synthesized molecules, pregnane-oximino-amino-alkyl-ether (compound 14b) improved glucose clearance in sucrose-loaded rats and exerted antihyperglycemic activity on STZ-induced diabetic rats. Further evaluation in genetically diabetic db/db mice showed temporal decrease in blood glucose, and improvement in glucose tolerance and lipid parameters, associated with mild improvement in the serum insulin level. Moreover, compound 14b treatment displayed an anti-dyslipidemic effect characterized by significant improvement in altered lipid parameters of the high-fructose, HFD-fed dyslipidemic hamster model. In vitro analysis in the cellular system suggested that compound 14b decreased glucose production in liver cells and stimulated glucose utilization in skeletal muscle cells. These beneficial effects of compound 14b were associated with the activation of the G-protein-coupled bile acid receptor TGR5. CONCLUSION: Compound 14b exhibits antidiabetic and anti-dyslipidemic activities through activating the TGR5 receptor system and can be developed as a lead for the management of type II diabetes and related metabolic complications.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Dyslipidemias/drug therapy , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Pregnanes/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Blood Glucose/drug effects , Cell Line , Cricetinae , Diabetes Mellitus, Experimental/metabolism , Dyslipidemias/metabolism , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Glucose Transporter Type 4/metabolism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/pharmacokinetics , Hypolipidemic Agents/therapeutic use , Male , Mice , Muscle, Skeletal/drug effects , Pregnanes/chemistry , Pregnanes/pharmacokinetics , Pregnanes/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
3.
Life Sci ; 289: 120221, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34902437

ABSTRACT

AIM: Free fatty acid-mediated obesity plays a crucial role in the pathogenesis of Type 2 Diabetes. FFA induced JNK activation acts as a central regulator in causing hepatic insulin resistance. Similarly, Pancreastatin, a chromogranin A peptide, serves as a crucial link between FFA-induced insulin resistance. Therefore, in the present work, we sought to test Pancreastatin inhibitor PSTi8 to ameliorate FFA-induced hepatic insulin resistance in in vitro and in vivo models. MATERIAL AND METHODS: To verify our objective, we exposed hepatocytes (HepG2 cells) with palmitate (0.3 mM) or palmitate + PSTi8 (200 nM). Parallelly mice were fed either HFD or HFD + PSTi8 (1 mg/kg). After 21 days animals were scanned for increased fat mass, along with GTT, ITT and PTT experiment to check glucose, and insulin tolerance. Furthermore, ROS generation and hepatic glycogen content was measured in FFA exposed hepatocytes. Gene expression and protein expression studies were further conducted to delineate the action mechanism of PSTi8. KEY FINDINGS: PSTi8 exposure decreased ROS accumulation, lipid accumulation, and reduced glycogen content in FFA-induced groups. It also enhances glucose uptake and reduces gluconeogenesis to combat the FFA effect. Furthermore, gene expression studies indicate that PSTi8 treatment reduces NADPH oxidase3 (NOX3) expression and inhibits JNK signaling, a predominant source of ROS-induced insulin resistance. SIGNIFICANCE: To summarize, the protective effect of PSTi8 on FFA-induced insulin resistance is mediated via inhibition of JNK signaling, which leads to decreased ROS generation and enhanced insulin sensitivity. Hence PSTi8 could be a therapeutic molecule to prevent western diet-induced insulin resistance.


Subject(s)
Chromogranin A/antagonists & inhibitors , Fatty Acids/metabolism , Insulin Resistance , MAP Kinase Kinase 4/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Chromogranin A/metabolism , Hep G2 Cells , Humans , Male , Mice
4.
Heliyon ; 6(10): e05133, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33033766

ABSTRACT

In the preceding study, we delineated that high-fat diet (HFD) consumption in mice increases the circulatory level of pancreastatin (PST), which additionally enhances the free fatty acid (FFA) concentration in circulation. Consequently, the aggravated FFA activates Fetuin-A, which facilitates hepatic lipid accumulation, insulin resistance (IR), and culminates in type 2 diabetes (T2D). Metformin (Met) is a widely known first-line drug for the treatment of T2D. We previously unveiled PSTi8, an inhibitor of PST, comprising antidiabetic property. Hence, we hypothesized that combination therapy of Met and PSTi8, at reduced therapeutic doses, would mitigate HFD-induced IR by inhibiting hepatic Fetuin-A in mice model of T2D. C57BL/6 mice were fed HFD for 12 weeks, followed by treatment with Met, PSTi8, and its combination for 10 days. Glucose and insulin tolerance tests were conducted. Circulatory levels of PST, Fetuin-A, and lipid markers were determined. Also, the mRNA and protein expression of Fetuin-A was assessed by qPCR, western blotting, and immunofluorescence. Moreover, the energy expenditure was measured by comprehensive laboratory animal monitoring system (CLAMS). Combination therapy displayed improved PST, Fetuin-A, and lipid profile in plasma. We also found reduced hepatic Fetuin-A, which reduced inhibitory phosphorylation of IRS and increased phosphorylation of AKT. Consequently, ameliorated hepatic lipogenesis, gluconeogenesis, and inflammation. Also, combination treatment attenuated Fetuin-A expression, lipid accumulation, and glucose production in palmitate-induced HepG2 cells. Altogether current study promulgates the beneficial effect of combination therapy of Met and PSTi8 (comparable to alone higher therapeutic doses) to ameliorate Fetuin-A activation, hepatic lipid accumulation, insulin resistance, and associated progressive pathophysiological alterations in T2D.

5.
Eur J Pharmacol ; 881: 173204, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32439261

ABSTRACT

Pancreastatin (PST), a chromogranin A (CHGA) derived peptide connects obesity with insulin resistance by inducing inflammation. Previously, we have evaluated potential activity of PST inhibitor (PSTi8) in liver and adipose tissue in type 2 diabetic mice model. In this study we further explore the therapeutic effect of PSTi8 on glucose metabolism in skeletal muscle cells/tissue and its effect on energy homeostasis in diet induced diabetic mice model. In in-vitro studies, we found that PSTi8 increases glucose uptake via enhanced GLUT4 translocation in L6 cells. This positive effect of PSTi8 led us to proceed with in-vivo studies in diabetic mice. C57BL/6 mice were fed HFD or HFrD diet for 12 weeks along with single STZ induction at 4th week followed by PSTi8 treatment. We found that HFD and HFrD model showed increased fat mass, caused glucose intolerance and insulin resistance, with accompanying proinflammatory effect on epididymal white adipose tissue (eWAT) together leading to skeletal muscle insulin resistance. Administration of PSTi8 protects from diet induced inflammatory response and enhances glucose tolerance and insulin sensitivity. PSTi8 improves circulating adipokine and lipid parameters, along with switch in macrophage polarisation from M1 to M2 in stromal vascular fraction of adipose tissue. In addition, treatment of PSTi8 also improves energy homeostasis, decreases circulatory non-esterified fatty acids level and inhibits ceramide deposition in muscle tissue. Overall this increased muscle insulin sensitivity is mediated via AKT/AS160/GLUT4 pathway activation. Our results reveal that PSTi8 inhibits the obesity mediated inflammation which enhances glucose disposal in skeletal muscle.


Subject(s)
Blood Glucose/drug effects , Chromogranin A/antagonists & inhibitors , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Insulin Resistance , Muscle, Skeletal/drug effects , Obesity/drug therapy , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/physiopathology , Adiposity/drug effects , Animals , Biomarkers/blood , Blood Glucose/metabolism , Chromogranin A/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Energy Metabolism/drug effects , GTPase-Activating Proteins/metabolism , Glucose Transporter Type 4/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Obesity/complications , Obesity/metabolism , Obesity/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Streptozocin , THP-1 Cells
6.
J Pharm Biomed Anal ; 180: 113074, 2020 Feb 20.
Article in English | MEDLINE | ID: mdl-31891874

ABSTRACT

PSTi8 is a 21 amino acid pancreastatin inhibitory peptide that demonstrated potent antidiabetic activity in insulin resistant rodent models. The goal of the current work is to establish and validate the LC-ESI-MS/MS bioanalytical assay of PSTi8 in mice plasma in order to unveil its pharmacokinetic (PK) behaviour for the first time. The MS detection of PSTi8 and diprotin A (internal standard, IS) was conducted with Q1/Q3 SRM transitions at 607.80 ([M+4 H]4+)/771.20 and 342.20/229.10, respectively using positive ESI. Phenomenex Aqua 5µ 125A (250 × 4.6 mm) column was utilized to separate PSTi8 and IS with a mobile phase consists of MeOH-0.1 % formic acid (1:1, v/v) using 0.4 mL/min flow rate. SPE using medium anion exchange cartridge (Oasis MAX) was used for the extraction of analyte and IS from the mice plasma and the extraction recovery was found to be >55 %. PSTi8 displayed good linearity across the 5-1000 ng/mL concentrations range. The intra- and inter- day accuracy was observed between 99.44-110.20 % and 99.66-110.93 %, respectively. The intra- and inter- day precision was observed between 2.61-4.03 % and 2.90-7.16 %, respectively. The intra-day and inter-day accuracy and precision data was within the 100 ±â€¯15 % nominal values recommended by the United States Food and Drug Administration bioanalytical guidance. The LC-MS/MS assay was validated effectively to investigate the PSTi8 plasma concentrations following intravenous and intraperitoneal PK studies in mice. The absolute bioavailability of PSTi8 was 52.74 ±â€¯13.50 %.


Subject(s)
Biological Assay/methods , Drug Development , Hypoglycemic Agents/blood , Animals , Biological Assay/instrumentation , Biological Availability , Calibration , Chromatography, Liquid , Drug Stability , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Injections, Intraperitoneal , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Reference Standards , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
7.
Heliyon ; 6(12): e05826, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33426334

ABSTRACT

AIMS: Hepatic steatosis in women confronting menopause is the manifestation of substantial fructose consumption and forms a positive feedback loop to develop endoplasmic reticulum (ER) stress. Previously pancreastatin inhibitor peptide-8 (PSTi8) and Metformin (Met) combination effectively ameliorated hepatic lipid accumulation in high fructose diet (HFrD) fed diabetic mice models at reduced doses. Moreover, SIRT-1 plays a crucial role in the regulation of SREBP-1c. Hence we hypothesized that Met and PSTi8 in combination (at therapeutic lower doses) could mitigate hepatic steatosis linked ER stress by activating SIRT-1 and precluding SREBP-1c in HFrD fed 4-Vinylcyclohexenediepoxide (HVCD) induced perimenopausal rats. MAIN METHODS: HVCD rats were fed HFrD for 12 weeks, accompanied by 14 days of treatment with Met, PSTi8, and combination. We confirmed model establishment by estrus cycle study, estradiol level, and intraperitoneal glucose tolerance test. Plasma lipid profile and liver function were determined. Also, mRNA and protein expressions were examined. Moreover, distribution of SIRT-1 and SREBP-1c was detected in HepG2 cells by immunofluorescence staining. KEY FINDINGS: HVCD group displayed augmented insulin resistance (IR), lipogenesis, and ER stress in the liver. Combination therapy improved the estrus cyclicity, estradiol, and lipid profile of HVCD rats. Met and PSTi8 combination reduced hepatic SREBP-1c and triggered SIRT-1 expression in high fructose-induced insulin-resistant HepG2 cells; consequently, combination therapy attenuated ER stress. SIGNIFICANCE: Succinctly, present research promotes impetus concerning the remedial impact of Met with PSTi8 at lower therapeutic doses to ameliorate hepatic IR, steatosis, and associated ER stress by revamping the SIRT-1/SREBP-1c axis in perimenopausal rats.

8.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1130-1131: 121829, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31670104

ABSTRACT

S011-2111 is a semicarbazone and chalcone hybrid demonstrating antiproliferative tumor cell-selective effects along with unique antimetastatic potential by mitigating PP2A-ß-catenin signalling pathway. The present study envisaged to explore the in vitro and in vivo pharmacokinetics of S011-2111. A sensitive and selective liquid chromatography-tandem mass spectrometry bioanalytical method was developed and validated to determine S011-2111. It has high permeability across intestinal membrane as observed in in situ single-pass intestinal perfusion study. It has high plasma protein binding and poor aqueous solubility. It was rapidly partitioning into plasma of blood, where it was moderately stable. In mice liver microsomal stability study, S011-2111 was stable against cytochrome P450 enzymes but undergoes rapid glucuronidation with intrinsic clearance of 148.6 ±â€¯48.3 µL/min/mg. Following 100 mg/kg oral dosing of S011-2111, the compound was detectable in the plasma samples up to 24 h with a maximum plasma concentration of 45 ±â€¯16.5 ng/mL at 2.4 ±â€¯0.1 h and absolute bioavailability of 1.68%. Knowledge from this research will assist in further development of S011-2111 as an anti-cancer agent.


Subject(s)
Chromatography, Liquid/methods , Enzyme Inhibitors , Protein Phosphatase 2/antagonists & inhibitors , Tandem Mass Spectrometry/methods , beta Catenin/antagonists & inhibitors , Animals , Enzyme Inhibitors/blood , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Erythrocytes , Female , Intestinal Absorption , Male , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization/methods
9.
Eur J Pharmacol ; 864: 172723, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31586632

ABSTRACT

Pancreastatin (PST), a chromogranin A derived peptide has anti-insulin effects and plays a significant role in obesity-induced insulin resistance. In obesity and type 2 diabetes mellitus, both insulin and PST level are elevated, but it is not clearly understood how anti-insulin effect of PST get regulated in hyperinsulinemic state. Simultaneously we have explored pancreastatin inhibitor PSTi8 against the native PST in the same hyperinsulinemic state. In in-vitro studies, we found that PST treatment increases lipid droplets and reactive oxygen species production in 3T3L1 adipocyte cells and theses effects of PST was found synergistic with chronic-insulin treatment. Treatment of PSTi8 in 3T3L1 adipocytes attenuates PST effect on lipid droplet formation and reactive oxygen species production. We further validated these findings in epididymal white adipose tissue of C57BL/6 mice, implanted with mini-osmotic insulin pump with and without PSTi8 for 4 weeks. We found that chronic hyperinsulinemia enhanced PST levels in circulation which in turn induces expression of various pro-inflammatory cytokines and oxidative stress. In addition, it also stimulated the expression of lipogenic genes, fat mass and body weight gain through the regulation of circulating adiponectin level. The change in PST mediated inflammatory and lipogenic parameters were attenuated by PSTi8 treatment, leading to enhanced insulin sensitivity and improved glucose homeostasis. PSTi8 rescue from PST mediated insulin resistance in adipose via inhibition of MAPK and NOX3-JNK stress signalling pathway which stimulates GLUT4 expression through activation of AKT-AS160 pathway. Thus PSTi8 may be a novel therapeutic agent for the treatment of hyperinsulinemia induced obesity and inflammation mediated insulin resistance.


Subject(s)
Chromogranin A/antagonists & inhibitors , Hyperinsulinism/complications , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , NADPH Oxidases/metabolism , Obesity/drug therapy , 3T3-L1 Cells , Animals , Homeostasis/drug effects , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Lipids/blood , Lipogenesis/drug effects , Male , Mice , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects
10.
Peptides ; 120: 170147, 2019 10.
Article in English | MEDLINE | ID: mdl-31473204

ABSTRACT

Increase in the prevalence of insulin resistance (IR) in peri-/post-menopause women is mainly due to hormone deficiency and lifestyle. PSTi8 (PEGKGEQEHSQQKEEEEEMAV-amide) is a pancreastatin inhibitor peptide which showed potent antidiabetic activity in genetic and lifestyle induced type 2 diabetic mice. In the present work, we have investigated the antidiabetic activity of PSTi8 in rat models of peri-/post-menopausal IR. 4-vinylcyclohexenediepoxide treated and ovariectomized rats were fed with high fat diet for 12 weeks to develop the peri-/post-menopausal IR. PSTi8 peptide was administered after the development of peri-/post-menopausal IR rats. PSTi8 (1 mg/kg, i.p) improved the glucose homeostasis which is characterized by elevated glycogenesis, enhanced glycolysis and reduced gluconeogenesis. PSTi8 suppressed palmitate- and PST- induced IR in HepG2 cells. PSTi8 treatment enhanced energy expenditure in peri-/post-menopausal IR rats. PSTi8 treatment increased insulin sensitivity in peri-/post-menopausal IR rats, may be mediated by modulating IRS1-2-phosphatidylinositol-3-kinase-AKT-GSK3ß and IRS1-2-phosphatidylinositol-3-kinase-PKCλ/ζ-SREBP1c signaling pathways in the liver. PSTi8 can act as a potential therapeutic peptide for the treatment of peri-/post-menopausal IR.


Subject(s)
Chromogranin A/antagonists & inhibitors , Dietary Fats/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Insulin Resistance , Isoenzymes/metabolism , Molecular Chaperones/metabolism , Peptides/pharmacology , Postmenopause/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Chromogranin A/metabolism , Female , Humans , Rats
11.
J Nutr Biochem ; 71: 35-44, 2019 09.
Article in English | MEDLINE | ID: mdl-31272030

ABSTRACT

Increased fructose intake has been linked to the development of dyslipidemia, obesity and impaired glucose tolerance. Due to its specific metabolic fate, fructose impairs normal lipid and carbohydrate metabolism and facilitates the non-enzymatic glycation reaction leading to enhanced accumulation of advanced glycation end products (AGEs). However, the formation of fructose-AGEs under in vivo setup and its tissue specific accumulation is less explored. Here, we investigated the impact of high fructose on AGEs accumulation in skeletal muscle and its causal role in impaired glucose homeostasis. In L6 rat skeletal muscle cells, chronic exposure to fructose induced AGEs accumulation and the cellular level of the receptor for AGEs (RAGE) and the effect was prevented by pharmacological inhibition of glycation. Under in vivo settings, Sprague Dawley rats exposed to 20% fructose in drinking water for 16 weeks, displayed increased fasting glycemia, impaired glucose tolerance, decreased skeletal muscle Akt (Ser-473) phosphorylation, and enhanced triglyceride levels in serum, liver and gastrocnemius muscle. We also observed a high level of AGEs in serum and gastrocnemius muscle of fructose-supplemented animals, associated with methylglyoxal accumulation and up regulated expression of RAGE in gastrocnemius muscle. Treatment with aminoguanidine inhibited fructose-induced AGEs accumulation and normalized the expression of RAGE and Dolichyl-Diphosphooligosaccharide-Protein Glycosyltransferase (DDOST) in gastrocnemius muscle. Inhibition of AGEs-RAGE axis counteracted fructose-mediated glucose intolerance without affecting energy metabolism. These data reveal diet-derived AGEs accumulation in skeletal muscle and the implication of tissue specific AGEs in metabolic derangement, that may open new perspectives in pathogenic mechanisms and management of metabolic diseases.


Subject(s)
Fructose/adverse effects , Glucose/metabolism , Glycation End Products, Advanced/metabolism , Muscle, Skeletal/drug effects , Receptor for Advanced Glycation End Products/metabolism , Animals , Cells, Cultured , Cytokines/blood , Energy Metabolism/drug effects , Glucose Intolerance , Homeostasis/drug effects , Inflammation/etiology , Lipid Metabolism/drug effects , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects
12.
Biomed Pharmacother ; 116: 108959, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31108350

ABSTRACT

AIMS: To investigate the role of pancreastatin inhibitor (PSTi8) in lipid homeostasis and insulin sensitivity in dexamethasone induced fatty liver disease associated type 2 diabetes. MAIN METHODS: Glucose releases assay, lipid O staining and ATP/AMP ratio were performed in HepG2 cells. Twenty four mice were randomly divided into 4 groups: Control group (saline), DEX (1 mg/kg, im) for 17 days, DEX+PSTi8 (acute 5 mg/kg and chronic 2 mg/kg, ip) for 10 days. The glucose, insulin and pyruvate tolerance tests (GTT, ITT and PTT), biochemical parameters and Oxymax-CLAMS were performed. Further to elucidate the action mechanisms of PSTi8, we performed genes expression and western blotting of biological samples. KEY FINDINGS: We found that PSTi8 suppresses hepatic glucose release, lipid deposition, oxidative stress induced by DEX, stimulates the cellular energy level in hepatocytes and enhances GRP78 activity. It reduces lipogensis and enhances fatty acid oxidation to improve insulin sensitivity and glucose tolerance in DEX induced diabetic mice. The above cellular effects are the result of activated AMPK signalling pathway in liver, which increases Srebp1c and ACC phosphorylation. The increased ACC phosphorylation suppresses protein kinase C activity and enhances insulin sensitivity. The increased expression of UCP3 in liver elicits fatty acid oxidation and energy expenditure, which suppress oxidative stress. SIGNIFICANCE: Thus the activation of AMPK signalling through GRP78, improves lipid homeostasis, enhances insulin sensitivity via inhibition of PKC activity. PSTi8 suppresses inflammation associated with incomplete fatty acid oxidation. Hence, PSTi8 may be a potential therapeutic agent to treat glucocorticoid-induced fatty liver associated type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chromogranin A/antagonists & inhibitors , Fatty Liver/enzymology , Fatty Liver/pathology , Heat-Shock Proteins/metabolism , Signal Transduction , Adenosine Triphosphate/metabolism , Adipokines/metabolism , Adiposity/drug effects , Animals , Chromogranin A/metabolism , Dexamethasone , Endoplasmic Reticulum Chaperone BiP , Energy Metabolism , Fatty Liver/blood , Glucose/metabolism , Hep G2 Cells , Homeostasis/drug effects , Humans , Insulin/blood , Insulin Resistance , Lipid Metabolism/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Tissue Distribution/drug effects
13.
J Pharm Biomed Anal ; 162: 205-214, 2019 Jan 05.
Article in English | MEDLINE | ID: mdl-30265980

ABSTRACT

S012-1332 is the first DNA ligase I inhibitor that demonstrated in vivo anti-breast cancer activity. The present study aimed to assess the in vivo pharmacokinetics of S012-1332 in rats and interpret them with in vitro findings. A sensitive and selective liquid chromatography-tandem mass spectrometry bioanalytical method was developed and validated to determine S012-1332. Following oral administration, the absolute bioavailability was 7.04%. The absorption was prolonged which can be explained by low solubility in simulated gastric fluid and several folds higher solubility in simulated intestinal fluid. The effective permeability across the intestinal membrane in in situ single pass perfusion study for S012-1332 was 5.58 ± 1.83 * 10-5 cm/sec compared to 5.99 ± 0.65 * 10-5 cm/sec for carbamazepine, with no significant difference, indicating S012-1332 has high permeability. It was rapidly partitioning into plasma in blood, where it was stable. Plasma protein binding was moderate which may have attributed to the rapid distribution out of the vascular compartment. The pharmacokinetics of S012-1332 was characterized by extensive clearance as seen with rat liver and intestinal microsomes. In vitro results elucidate the in vivo pharmacokinetic data. These findings provide crucial information for further development of S012-1332 as anti-breast cancer agent.


Subject(s)
Antineoplastic Agents/pharmacokinetics , DNA Ligase ATP/antagonists & inhibitors , Enzyme Inhibitors/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Biological Availability , Chromatography, Liquid , DNA Ligase ATP/metabolism , Drug Stability , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Inactivation, Metabolic , Intestinal Absorption , Intestinal Mucosa/metabolism , Male , Microsomes, Liver/metabolism , Permeability , Protein Binding , Rats, Sprague-Dawley , Reproducibility of Results , Solubility , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
14.
Drug Dev Res ; 79(7): 339-351, 2018 11.
Article in English | MEDLINE | ID: mdl-30284738

ABSTRACT

Preclinical Research & Development Withanolide A (WA), a steroidal lactone is a major bioactive constituent of Withania somnifera (L.) with remarkable neuropharmacological activity. In this study, we investigated the permeability, plasma protein binding (PPB), blood partitioning, intravenous (i.v.), and oral pharmacokinetics as well as i.v. tissue distribution (TD) of pure WA in a rat model. The PPB, RBCs partitioning, and permeability of WA were determined by Ultra Performance Liquid Chromatography (UPLC) method. However, the pharmacokinetics and TD of WA were evaluated by validated and sensitive liquid chromatography coupled mass spectrometry (LC-ESI-MS/MS) method. The PPB and permeability of WA were determined by equilibrium dialysis and parallel artificial membrane permeability assay method, respectively. The results demonstrated that WA has high PPB and passive permeability. Furthermore, WA was found to have fast equilibration between RBCs and plasma. Following i.v. (2 mg/kg) and per-oral (25 mg/kg) administration of WA, the max concentration (Cmax ) in plasma was found as 85.53 ± 6.54 and 48.04 ±5.78 ng/mL, respectively. The TD study results indicated that WA has a rapid and wide TD. The maximum concentration in various tissues was found in following order: Clung > Cliver > Ckidney ≈ Cspleen > Cheart > Cbrain . The preclinical in vitro, as well as pharmacokinetics and TD results, are anticipated to support the future preclinical and clinical application of WA.


Subject(s)
Blood Proteins/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Phytosterols/pharmacokinetics , Withania , Withanolides/pharmacokinetics , Animals , Blood Proteins/analysis , Lactones/analysis , Lactones/blood , Lactones/pharmacokinetics , Male , Neuroprotective Agents/analysis , Neuroprotective Agents/blood , Permeability/drug effects , Phytosterols/analysis , Phytosterols/blood , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry/methods , Tissue Distribution/drug effects , Tissue Distribution/physiology , Withanolides/analysis , Withanolides/blood
15.
Sci Rep ; 8(1): 8715, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880906

ABSTRACT

Pancreastatin (PST) is an endogenous peptide which regulates glucose and lipid metabolism in liver and adipose tissues. In type 2 diabetic patients, PST level is high and plays a crucial role in the negative regulation of insulin sensitivity. Novel therapeutic agents are needed to treat the diabetes and insulin resistance (IR) against the PST action. In this regard, we have investigated the PST inhibitor peptide-8 (PSTi8) action against diabetogenic PST. PSTi8 rescued PST-induced IR in HepG2 and 3T3L1 cells. PSTi8 increases the GLUT4 translocation to cell surface to promote glucose uptake in L6-GLUT4myc cells. PSTi8 treatment showed an increase in insulin sensitivity in db/db, high fat and fructose fed streptozotocin (STZ) induced IR mice. PSTi8 improved the glucose homeostasis which is comparable to metformin in diabetic mice, characterized by elevated glucose clearance, enhanced glycogenesis, enhanced glycolysis and reduced gluconeogenesis. PST and PSTi8 both were docked to the GRP78 inhibitor binding site in protein-protein docking, GRP78 expression and its ATPase activity studies. The mechanism of action of PSTi8 may be mediated by activating IRS1/2-phosphatidylinositol-3-kinase-AKT (FoxO1, Srebp-1c) signaling pathway. The discovery of PSTi8 provides a promising therapeutic agent for the treatment of metabolic diseases mainly diabetes.


Subject(s)
Chromogranin A/antagonists & inhibitors , Diabetes Mellitus, Experimental/drug therapy , Insulin Resistance , Peptides , 3T3-L1 Cells , Animals , Chromogranin A/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Endoplasmic Reticulum Chaperone BiP , Gluconeogenesis/drug effects , Glucose/metabolism , Glycolysis/drug effects , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , Male , Mice , Molecular Docking Simulation , Peptides/chemistry , Peptides/pharmacology , Protein Domains , Signal Transduction/drug effects
16.
Free Radic Res ; 52(5): 568-582, 2018 May.
Article in English | MEDLINE | ID: mdl-29544378

ABSTRACT

Oxidative stress due to enhanced production or reduced scavenging of reactive oxygen species (ROS) has been associated with diet (dyslipidemia) induced obesity and insulin resistance (IR). The present study was undertaken to assess the role of p47phox in IR using wild type (WT) and p47phox-/- mice, fed with different diets (HFD, LFD or Chow). Augmented body weight, glucose intolerance and reduced insulin sensitivity were observed in p47phox-/- mice fed with 45% HFD and 10% LFD. Further, body fat and circulating lipids were increased significantly with 5 weeks LFD feeding in p47phox-/- mice, while parameters of energy homeostasis were reduced as compared with WT mice. LFD fed knockout (KO) mice showed an enhanced hepatic glycogenolysis, and reduced insulin signalling in liver and adipose tissue, while skeletal muscle tissue remained unaffected. A significant increase in hepatic lipids, adiposity, as well as expression of genes regulating lipid synthesis, breakdown and efflux were observed in LFD fed p47phox-/- mice after 5 weeks. On the other hand, mice lacking p47phox demonstrated altered glucose tolerance and tissue insulin sensitivity after 5 weeks chow feeding, while changes in body weight, respiratory exchange ratio (RER) and heat production are non-significant. Our data demonstrate that lack of p47phox is sufficient to induce IR through altered glucose and lipid utilization by the liver and adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Dyslipidemias/metabolism , Glucose/metabolism , Insulin Resistance , Liver/metabolism , NADPH Oxidases/genetics , Obesity/metabolism , Adipose Tissue/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Diet, Fat-Restricted , Diet, High-Fat , Dyslipidemias/etiology , Dyslipidemias/genetics , Dyslipidemias/pathology , Gene Expression Regulation , Glycogenolysis/genetics , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Lipid Metabolism/genetics , Liver/pathology , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , NADPH Oxidases/deficiency , Obesity/etiology , Obesity/genetics , Obesity/pathology , Oxidative Stress , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
17.
Mol Cell Endocrinol ; 460: 134-151, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28736255

ABSTRACT

Augmenting glucose utilization and energy expenditure in skeletal muscle via AMP-activated protein kinase (AMPK) is an imperative mechanism for the management of type 2 diabetes. Chemical derivatives (2a-2h, 3, 4a-4d, 5) of the isoalantolactone (K007), a bioactive molecule from roots of Inula racemosa were synthesized to optimize the bioactivity profile to stimulate glucose utilization in skeletal muscle cells. Interestingly, 4a augmented glucose uptake, driven by enhanced translocation of glucose transporter 4 (GLUT4) to cell periphery in L6 rat skeletal muscle cells. The effect of 4a was independent to phosphatidylinositide-3-kinase (PI-3-K)/Akt pathway, but mediated through Liver kinase B1 (LKB1)/AMPK-dependent signaling, leading to activation of downstream targets acetyl coenzyme A carboxylase (ACC) and sterol regulatory element binding protein 1c (SREBP-1c). In db/db mice, 4a administration decreased blood glucose level and improved body mass index, lipid parameters and glucose tolerance associated with elevation of GLUT4 expression in skeletal muscle. Moreover, 4a increased energy expenditure via activating substrate utilization and upregulated the expression of thermogenic transcription factors and mitochondrial proteins in skeletal muscle, suggesting the regulation of energy balance. These findings suggest the potential implication of isoalantolactone derivatives for the management of diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Experimental/metabolism , Energy Metabolism/drug effects , Glucose/metabolism , Muscle, Skeletal/metabolism , Sesquiterpenes/pharmacology , Signal Transduction , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Rats , Sesquiterpenes/chemistry , Time Factors
19.
Sci Rep ; 7: 41009, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28106120

ABSTRACT

On the basis of diet induced obesity and KO mice models, nitric oxide is implied to play an important role in the initiation of dyslipidemia induced insulin resistance. However, outcomes using iNOS KO mice have so far remained inconclusive. The present study aimed to assess IR in iNOS KO mice after 5 weeks of LFD feeding by monitoring body composition, energy homeostasis, insulin sensitivity/signaling, nitrite content and gene expressions changes in the tissues. We found that body weight and fat content in KO mice were significantly higher while the respiratory exchange ratio (RER), volume of carbon dioxide (VCO2), and heat production were lower as compared to WT mice. Furthermore, altered systemic glucose tolerance, tissue insulin signaling, hepatic gluconeogenesis, augmented hepatic lipids, adiposity, as well as gene expression regulating lipid synthesis, catabolism and efflux were evident in iNOS KO mice. Significant reduction in eNOS and nNOS gene expression, hepatic and adipose tissue nitrite content, circulatory nitrite was also observed. Oxygen consumption rate of mitochondrial respiration has remained unaltered in KO mice as measured using extracellular flux analyzer. Our findings establish a link between the NO status with systemic and tissue specific IR in iNOS KO mice at 5 weeks.


Subject(s)
Adipose Tissue/physiopathology , Glucose/metabolism , Homeostasis , Insulin Resistance , Lipid Metabolism , Liver/physiopathology , Nitric Oxide Synthase Type II/deficiency , Animals , Body Fat Distribution , Body Weight , Diet/methods , Mice, Knockout , Oxygen Consumption , Respiration , Thermogenesis
20.
Medchemcomm ; 8(2): 329-337, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-30108748

ABSTRACT

Based on high throughput screening of our chemical library, we identified two 4,5-dihydro-2H-benzo[e]indazole derivatives (5d and 5g), which displayed a significant effect on glucose uptake in L6 skeletal muscle cells. Based on these lead molecules, a series of benzo[e]indazole derivatives were prepared. Among all the synthesized dihydro-2H-benzo[e]indazoles, 8-(methylthio)-2-phenyl-6-p-tolyl-4,5-dihydro-2H-benzo[e]indazole-9-carboxylate (5e) showed significant glucose uptake stimulation in L6 skeletal muscle cells, even better than lead compounds. Additionally, 5e decreased glucagon-induced glucose release in HepG2 hepatoma cells. The 2H-benzo[e]indazole 5e exerted an antihyperglycemic effect in normal, sucrose challenged streptozotocin-induced diabetic rats and type 2 diabetic db/db mice. Treatment with 5e at a dose of 30 mg kg-1 in db/db mice caused a significant decrease in triglyceride and total cholesterol levels and increased the HDL-C level in a significant manner. The mechanistic studies revealed that the 2H-benzo[e]indazole 5e significantly stimulated insulin-induced signaling at the level of IRS-1, Akt and GSK-3ß in L6 skeletal muscle cells, possibly by inhibiting protein tyrosine phosphatase-1B. This new 2H-benzo[e]indazole derivative has potential for the treatment of diabetes with improved lipid profile.

SELECTION OF CITATIONS
SEARCH DETAIL
...