Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Mol Cell Cardiol ; 158: 140-152, 2021 09.
Article in English | MEDLINE | ID: mdl-34081950

ABSTRACT

3-Hydroxy-3-methyl glutaryl-coenzyme A reductase (Hmgcr) encodes the rate-limiting enzyme in the cholesterol biosynthesis pathway. The regulation of Hmgcr in rat models of genetic hypertension (viz. Spontaneously Hypertensive Rat [SHR] and its normotensive control Wistar/Kyoto [WKY] strain) is unclear. Interestingly, Hmgcr transcript and protein levels are diminished in liver tissues of SHR as compared to WKY. This observation is consistent with the diminished plasma cholesterol level in SHR animals. However, the molecular basis of these apparently counter-intuitive findings remains completely unknown. Sequencing of the Hmgcr promoter in SHR and WKY strains reveals three variations: A-405G, C-62T and a 11 bp insertion (-398_-388insTGCGGTCCTCC) in SHR. Among these variations, A-405G occurs at an evolutionarily-conserved site among many mammals. Moreover, SHR-Hmgcr promoter displays lower activity than WKY-Hmgcr promoter in various cell lines. Transient transfections of Hmgcr-promoter mutants and in silico analysis suggest altered binding of Runx3 and Srebf1 across A-405G site. On the other hand, C-62T and -398_-388insTGCGGTCCTCC variations do not appear to contribute to the reduced Hmgcr promoter activity in SHR as compared to WKY. Indeed, chromatin immunoprecipitation assays confirm differential binding of Runx3 and Srebf1 to Hmgcr promoter leading to reduced expression of Hmgcr in SHR as compared to WKY under basal as well as cholesterol-modulated conditions. Taken together, this study provides, for the first time, molecular basis for diminished Hmgcr expression in SHR animals, which may account for the reduced circulating cholesterol level in this widely-studied model for cardiovascular diseases.


Subject(s)
Alleles , Gene Expression Regulation , Gene Expression , Hydroxymethylglutaryl CoA Reductases/genetics , Hypertension/enzymology , Hypertension/genetics , Promoter Regions, Genetic/genetics , Animals , CHO Cells , Core Binding Factor Alpha 3 Subunit/genetics , Cricetulus , Female , HEK293 Cells , Hep G2 Cells , Humans , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sterol Regulatory Element Binding Protein 1/genetics , Transfection
2.
Regul Toxicol Pharmacol ; 123: 104960, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34022260

ABSTRACT

Cassia occidentalis Linn (CO) is an annual/perennial plant having traditional uses in the treatments of ringworm, gastrointestinal ailments and piles, bone fracture, and wound healing. Previously, we confirmed the medicinal use of the stem extract (ethanolic) of CO (henceforth CSE) in fracture healing at 250 mg/kg dose in rats and described an osteogenic mode of action of four phytochemicals present in CSE. Here we studied CSE's preclinical safety and toxicity. CSE prepared as per regulations of Current Good Manufacturing Practice for human pharmaceuticals/phytopharmaceuticals and all studies were performed in rodents in a GLP-accredited facility. In acute dose toxicity as per New Drug and Clinical Trial Rules, 2019 (prior name schedule Y), in rats and mice and ten-day dose range-finding study in rats, CSE showed no mortality and no gross abnormality at 2500 mg/kg dose. Safety Pharmacology showed no adverse effect on central nervous system, cardiovascular system, and respiratory system at 2500 mg/kg dose. CSE was not mutagenic in the Ames test and did not cause clastogenicity assessed by in vivo bone marrow genotoxicity assay. By a sub chronic (90 days) repeated dose (as per OECD, 408 guideline) study in rats, the no-observed-adverse-effect-level was found to be 2500 mg/kg assessed by clinico-biochemistry and all organs histopathology. We conclude that CSE is safe up to 10X the dose required for its osteogenic effect.


Subject(s)
Phytochemicals/toxicity , Plant Extracts/toxicity , Senna Plant , Animals , Ethanol , Mice , No-Observed-Adverse-Effect Level , Rats , Rodentia , Toxicity Tests
3.
J Vasc Res ; 54(4): 195-199, 2017.
Article in English | MEDLINE | ID: mdl-28618422

ABSTRACT

OBJECTIVE: Fluid-phase pinocytosis is a receptor-independent mechanism of endocytosis that occurs in all mammalian cells and may be a mechanism for the uptake of LDL by macrophages. As there are currently no methods for the measurement of fluid-phase pinocytosis by individual aortic cells in vivo, we sought to identify a suitable method. METHODS: ApoE-/- mice were retro-orbitally injected with AngioSPARK fluorescent nanoparticles specifically designed to not interact with cells. After 24 h, mice were sacrificed, and the aortas were isolated and then digested to analyze aortic cell uptake of AngioSPARK by flow cytometry. RESULTS: CD11b-expressing aortic macrophages from mice injected with AngioSPARK showed high levels of fluid-phase pinocytosis compared to aortic cells not expressing CD11b (4,393.7 vs. 408.3 mean fluorescence intensity [MFI], respectively). CONCLUSION: This new technique allows for the measurement of fluid-phase pinocytosis by aortic cells in vivo, making it possible to examine the cell-signaling molecules and drugs that affect this process. Published by S. Karger AG, Basel.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Flow Cytometry/methods , Macrophages/metabolism , Pinocytosis , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , CD11b Antigen/metabolism , Disease Models, Animal , Fluorescent Dyes/metabolism , Genetic Predisposition to Disease , Male , Mice, Knockout , Phenotype
4.
J Cardiovasc Pharmacol ; 69(5): 314-325, 2017 May.
Article in English | MEDLINE | ID: mdl-28207427

ABSTRACT

OBJECTIVE: Although atherosclerosis is described in New Zealand White rabbit's iliac artery, yet details of time-dependent atherosclerosis progression are not well known. Further, a well characterized accelerated model of atherosclerosis is also required for the screening of candidate drugs to target specific steps of atherosclerosis development. The present study extensively characterizes the time-dependent plaque composition and functional responses of the atherosclerosis in rabbit iliac artery and its modification by simvastatin. METHODS: Atherosclerosis was induced with a combination of balloon injury and atherogenic diet (AD) (1% cholesterol, 6% peanut oil) in rabbit's iliac artery. Atherosclerosis progression was evaluated on days 8, 10, 15, 21, 35, and 56 after AD feeding. The plaque characterization was done using histology, real-time reverse transcription-polymerase chain reaction, and vasoreactivity experiments. The standard anti-hyperlipidemic drug, simvastatin (5 mg·kg·d), was used to investigate its effect on atherosclerotic changes. RESULTS: Plasma lipids were elevated in a progressive manner after AD feeding from days 8 to 56. Similarly, arterial lipids, Monocyte Chemoattractant Protein-1 (MCP-1) level along with infiltration of macrophages in the lesion area were also increased from day 15 onward. This resulted in a significant increase in the plaque area and intimal-medial thickness ratio in contrast to normal animals. Inflammatory milieu was observed with a significant increase in expression of pro-inflammatory regulators like MCP-1, Tumor Necrosis Factor-α (TNF-α) and Vascular Cell Adhesion Molecule-1 (VCAM-1), whereas anti-inflammatory cytokine interleukin 10 decreased as disease progressed. Endothelial dysfunction was also observed, specifically Acetylcholine (ACh)-induced vasorelaxation was reduced from day 8 onward, whereas the phenylephrine-induced vasoconstriction response was progressively reduced from day 15 in the iliac artery. Ground substances including proteoglycans, α-actin, and collagen content along with metalloproteinase-9 and Tissue inhibitor of metalloproteinases-1 (TIMP-1) inhibitors were significantly augmented at later time points, day 21 onward. Simvastatin treatment for 35 days, at a dose having no significant effect on plasma lipid levels, significantly reduced atherosclerotic progression as evident by reduced macrophage content, inflammatory burden, and extracellular matrix component like proteoglycans and metalloproteinase-9. CONCLUSIONS: The authors observed that AD feeding with balloon injury in the rabbit iliac artery accelerated the progression of atherosclerosis and exhibited predominant features of type III human lesion within 8 weeks (56 days). Simvastatin treatment for 35 days exhibited anti-atherosclerotic efficacy without significantly lowering the circulating lipids. The current study thus provides an insight into the time-dependent atherosclerotic progression in rabbit iliac artery and highlights its utility for anti-atherosclerotic evaluation of the candidate drugs.


Subject(s)
Atherosclerosis/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Iliac Artery/drug effects , Plaque, Atherosclerotic , Simvastatin/pharmacology , Angioplasty, Balloon , Animals , Atherosclerosis/blood , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Disease Progression , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Extracellular Matrix/metabolism , Iliac Artery/metabolism , Iliac Artery/pathology , Iliac Artery/physiopathology , Inflammation Mediators/blood , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Male , Rabbits , Time Factors , Vascular Remodeling/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects
5.
Phytother Res ; 29(6): 910-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25851068

ABSTRACT

Azadirachta indica is well known medicinal plant mentioned in ancient herbal texts. It has been extensively used in Ayurvedic, Unani and Homoeopathic medicine and has become a luminary of modern medicine. As part of our drug discovery program we isolated azadiradione from the ethanolic extract of seeds of A. indica and evaluated for in-vivo antiulcer activity in cold restraint induced gastric ulcer model, aspirin induced gastric ulcer model, alcohol induced gastric ulcers model and pyloric ligation induced ulcer model. Azadiradione exhibited potent antiulcer activity through the inhibition of H+ K+-ATPase (proton pump) activity via its cytoprotective effect and also via its antisecretory effect. This combined effect has valuable potential in the future treatment of peptic ulceration.


Subject(s)
Anti-Ulcer Agents/pharmacology , Azadirachta/chemistry , Limonins/pharmacology , Plant Extracts/pharmacology , Stomach Ulcer/drug therapy , Animals , Anti-Ulcer Agents/isolation & purification , Dinoprostone/chemistry , Disease Models, Animal , Female , Limonins/isolation & purification , Male , Plants, Medicinal/chemistry , Proton Pump Inhibitors/isolation & purification , Proton Pump Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Seeds/chemistry
6.
J Cell Biochem ; 114(3): 532-40, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22961925

ABSTRACT

Neutrophils/polymorphonuclear leukocytes (PMNs), an important component of innate immune system, release extracellular traps (NETs) to eliminate invaded pathogens; however understanding of the role of signaling molecules/proteins need to be elucidated. In the present study role of p38 MAPK and extracellular signal regulated kinase (ERK) against phorbol 12-myristate 13-acetate (PMA) induced reactive oxygen species (ROS) generation and NETs formation has been investigated. Human neutrophils were treated with PMA to induce free radical generation and NETs release, which were monitored by NBT reduction and elastase/DNA release, respectively. PMA treatment led to the time dependent phosphorylation of p38 MAPK and ERK in PMNs. Pretreatment of PMNs with SB202190 or U0126 did not significantly reduce PMA induce free radical generation, but prevented NETs release. Pretreatment of PMNs with NADPH oxidase inhibitor (diphenyleneiodonium chloride) significantly reduced free radical generation, p38 MAPK and ERK phosphorylation as well as NETs release, suggesting that p38 MAPK and ERK activation was downstream to free radical generation. The present study thus demonstrates ROS dependent activation of ERK and p38 MAPK, which mediated PMA induced NETs release from human neutrophils.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Butadienes/pharmacology , Enzyme Activation , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neutrophils/drug effects , Nitriles/pharmacology , Onium Compounds/pharmacology , Phosphorylation , Pyridines/pharmacology , Tetradecanoylphorbol Acetate
7.
J Lipid Res ; 53(1): 34-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22058424

ABSTRACT

Accumulation of cholesterol by macrophage uptake of LDL is a key event in the formation of atherosclerotic plaques. Previous research has shown that granulocyte-macrophage colony-stimulating factor (GM-CSF) is present in atherosclerotic plaques and promotes aortic lipid accumulation. However, it has not been determined whether murine GM-CSF-differentiated macrophages take up LDL to become foam cells. GM-CSF-differentiated macrophages from LDL receptor-null mice were incubated with LDL, resulting in massive macrophage cholesterol accumulation. Incubation of LDL receptor-null or wild-type macrophages with increasing concentrations of ¹²5I-LDL showed nonsaturable macrophage LDL uptake that was linearly related to the amount of LDL added, indicating that LDL uptake was mediated by fluid-phase pinocytosis. Previous studies suggest that phosphoinositide 3-kinases (PI3K) mediate macrophage fluid-phase pinocytosis, although the isoform mediating this process has not been determined. Because PI3Kγ is known to promote aortic lipid accumulation, we investigated its role in mediating macrophage fluid-phase pinocytosis of LDL. Wild-type macrophages incubated with LDL and the PI3Kγ inhibitor AS605240 or PI3Kγ-null macrophages incubated with LDL showed an ∼50% reduction in LDL uptake and cholesterol accumulation compared with wild-type macrophages incubated with LDL only. These results show that GM-CSF-differentiated murine macrophages become foam cells by fluid-phase pinocytosis of LDL and identify PI3Kγ as contributing to this process.


Subject(s)
Cholesterol, LDL/metabolism , Class Ib Phosphatidylinositol 3-Kinase/physiology , Foam Cells/physiology , Macrophages/drug effects , Pinocytosis/drug effects , Animals , Cell Differentiation/drug effects , Foam Cells/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Lipoproteins, LDL , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Quinoxalines/pharmacology , Thiazolidinediones/pharmacology
8.
Cytometry A ; 81(3): 238-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22170804

ABSTRACT

Neutrophils expel extracellular traps (NETs) to entrap and exterminate the invaded micro-organisms. Acute/chronic inflammatory disorders are often observed with aberrantly enhanced NETs formation and high nitric oxide (NO) availability. Recent study from this laboratory demonstrated release of NETs from human neutrophils following treatment with SNP or SNAP. This study is an extension of our previous finding to explore the extracellular bacterial killing, source of DNA in the expelled NETs, their ability to induce proinflammatory cytokines release from platelets/THP-1 cells, and assessment of NO-mediated free radical formation by using a consistent NO donor, DETA-NONOate. NO-mediated NETs exhibited extracellular bacterial killing as determined by colony forming units. NO-mediated NETs formation was due to the activation of NADPH oxidase and myeloperoxidase. NO- or PMA-mediated NETs were positive for both nuclear and mitochondrial DNA as well as proteolytic enzymes. Incubation of NETs with human platelets enhanced the release of IL-1ß and IL-8, while with THP-1 cells, release of IL-1ß, IL-8, and TNFα was observed. This study demonstrates that NO by augmenting enzymatic free radical generation release NETs to promote extracellular bacterial killing. These NETs were made up of mitochondrial and nuclear DNA and potentiated release of proinflammatory cytokines.


Subject(s)
DNA, Mitochondrial/metabolism , DNA/metabolism , Inflammation/immunology , Neutrophil Activation , Neutrophils/cytology , Neutrophils/metabolism , Adult , Blood Platelets/metabolism , Free Radicals , Humans , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Mitochondria/genetics , NADPH Oxidases/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Peroxidase/metabolism , Tumor Necrosis Factor-alpha
9.
Hypertens Res ; 44(2): 154-167, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32917968

ABSTRACT

Kidneys have a high resting metabolic rate and low partial pressure of oxygen due to enhanced mitochondrial oxygen consumption and ATP production needed for active solute transport. Heightened mitochondrial activity leads to progressively increasing hypoxia from the renal cortex to the renal medulla. Renal hypoxia is prominent in hypertensive rats due to increased sodium reabsorption within the nephrons, which demands higher energy production by oxidative phosphorylation (OXPHOS). Consequently, spontaneously hypertensive rats (SHR) display greater oxygen deficiency (hypoxia) than normotensive Wistar Kyoto rats (WKY). Here, we sought to investigate the expression of key proteins for mitochondrial biogenesis in SHR and WKY, and study the regulation of mitochondrial transcription factors (mtTFs) under in vitro hypoxic conditions in renal epithelial cells. We report that renal expressions of hypoxia-inducible factor-1-alpha (HIF-1α), peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α), mtTFs, and OXPHOS proteins are elevated in SHR compared to WKY. In addition, our experiments in cultured kidney cells demonstrate that acute hypoxia augments the expression of these genes. Furthermore, we show that the transcripts of HIF-1α and mtTFs are positively correlated in various human tissues. We reveal, for the first time to our knowledge, that HIF-1α transactivates mtTF genes by direct interaction with their promoters in rat kidney epithelial cells (NRK-52E) under acute hypoxia. Concomitant increases in the mitochondrial DNA and RNA, and OXPHOS proteins are observed. Taken together, this study suggests that hypoxia within the renal epithelial cells may enhance mitochondrial function to meet the energy demand in proximal tubular cells during prehypertensive stages in kidneys of young SHR.


Subject(s)
Hypertension , Animals , Epithelial Cells , Hypoxia , Mitochondria , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Transcription Factors/genetics
10.
Mol Cell Biol ; 40(9)2020 04 13.
Article in English | MEDLINE | ID: mdl-32071155

ABSTRACT

Hypercholesterolemia is a strong predictor of cardiovascular diseases. The 3-hydroxy-3-methylglutaryl coenzyme A reductase gene (Hmgcr) coding for the rate-limiting enzyme in the cholesterol biosynthesis pathway is a crucial regulator of plasma cholesterol levels. However, the posttranscriptional regulation of Hmgcr remains poorly understood. The main objective of this study was to explore the role of microRNAs (miRNAs) in the regulation of Hmgcr expression. Systematic in silico predictions and experimental analyses reveal that miRNA 27a (miR-27a) specifically interacts with the Hmgcr 3' untranslated region in murine and human hepatocytes. Moreover, our data show that Hmgcr expression is inversely correlated with miR-27a levels in various cultured cell lines and in human and rodent tissues. Actinomycin D chase assays and relevant experiments demonstrate that miR-27a regulates Hmgcr by translational attenuation followed by mRNA degradation. Early growth response 1 (Egr1) regulates miR-27a expression under basal and cholesterol-modulated conditions. miR-27a augmentation via tail vein injection of miR-27a mimic in high-cholesterol-diet-fed Apoe-/- mice shows downregulation of hepatic Hmgcr and plasma cholesterol levels. Pathway and gene expression analyses show that miR-27a also targets several other genes (apart from Hmgcr) in the cholesterol biosynthesis pathway. Taken together, miR-27a emerges as a key regulator of cholesterol biosynthesis and has therapeutic potential for the clinical management of hypercholesterolemia.


Subject(s)
Cholesterol/biosynthesis , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Cholesterol/genetics , Cholesterol/metabolism , Databases, Genetic , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipogenesis/genetics , Liver/metabolism , Mice , MicroRNAs/genetics , RNA Stability , Rats , Transfection
11.
Curr Vasc Pharmacol ; 7(1): 75-109, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19149643

ABSTRACT

The recent failure of candidate drugs like cholesterol ester transfer protein (CETP) and acyl-CoA:cholesterol acyltransferase (ACAT) inhibitors calls for a revised approach for screening anti-atherosclerotic drugs and development of new models of atherosclerosis. For this it is important to understand the mechanism of the disease in a particular model. Models simultaneously showing hyperlipidemia, inflammation and associated complications of diabetes and hypertension will serve the purpose better as they mimic the actual clinical condition. Besides this, analyzing candidate molecules in vivo, in vitro and at various levels of atherosclerosis progression is important. Models based on various cells and process involved in atherosclerosis should be used for screening candidate molecules. The challenge lies in bridging the gap between genetically friendly small animal and human-like bigger animal models. Sequencing of the mouse and human genome, development of a single nucleotide polymorphism (SNP) database and in silico quantitative trait loci (QTL) linkage analysis may enhance the understanding of atherosclerosis and help develop new therapeutic targets.


Subject(s)
Arteriosclerosis/physiopathology , Disease Models, Animal , Drug Delivery Systems , Animals , Arteriosclerosis/drug therapy , Databases, Genetic , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Sequence Analysis, DNA/methods
12.
Cell Mol Immunol ; 16(12): 946, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31511637

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
J Mol Biol ; 431(6): 1127-1147, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30738894

ABSTRACT

Monoamine oxidase B (MAO-B), a flavoenzyme located in the outer mitochondrial membrane, is involved in the catabolism of monoamines. Altered levels of MAO-B are associated with cardiovascular/neuronal diseases. However, molecular mechanisms of MAO-B gene regulation are partially understood. We undertook a systematic analysis of the MAO-B gene to identify the key transcriptional/post-transcriptional regulatory molecules. Expression of MAO-B promoter-reporter constructs in cultured cells identified the -144/+25-bp domain as the core promoter region. Stringent in silico analysis of this core promoter predicted binding sites for several transcription factors. Over-expression/down-regulation of transcription factors Sp1/Egr1/CREB increased/decreased the MAO-B promoter-reporter activity and endogenous MAO-B protein level. Electrophoretic mobility shift assays and ChIP assays provided evidence for interactions of Sp1/Egr1/CREB with the MAO-B promoter. MAOB transcript level also positively correlated with the transcript level of Sp1/Egr1/CREB in various human tissue samples. Computational predictions using multiple algorithms coupled with systematic functional analysis revealed direct interactions of the microRNAs miR-1224 and miR-300 with MAO-B 3'-UTR. Dopamine dose-dependently enhanced MAO-B transcript and protein levels via increased binding of CREB to MAO-B promoter and reduced miR-1224/miR-300 levels. 8-Bromo-cAMP and forskolin augmented MAO-B expression, whereas inhibition of PKA diminished the gene expression suggesting involvement of cAMP-PKA axis. Interestingly, Sp1/Egr1/CREB/miR-1224 levels correlate with MAO-B expression in rodent models of hypertension/MPTP-induced neurodegeneration, indicating their roles in governing MAO-B gene expression in these disease states. Taken together, this study elucidates the previously unknown roles of the transcription factors Sp1/Egr1/CREB and microRNAs miR-1224/miR-300 in regulating MAO-B gene expression under basal/disease states involving dysregulated catecholamine levels.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Protein 1/metabolism , Gene Expression Regulation, Enzymologic , MicroRNAs/metabolism , Monoamine Oxidase/genetics , Sp1 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , Cricetulus , Down-Regulation , Genes, Reporter , Genetic Predisposition to Disease/genetics , Humans , Male , Mice , Monoamine Oxidase/metabolism , Promoter Regions, Genetic , Rats , Transcription Factors , Transcription, Genetic
14.
J Ethnopharmacol ; 213: 81-91, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29129602

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Xylocarpus moluccensis (Lamk.) M. Roem of family Meliaceae has triterpenoids rich fruits. Triterpenoids have been known to possess cardioprotection and anti-atherosclerotic activities (Han and Bakovic, 2015; Wu et al., 2009). Standardized fraction of these fruits exhibited anti-dyslipidemic (Srivastava et al., 2015), anti-inflammatory (Ravangpai et al., 2011) and CNS depressant activity (Sarker et al., 2007). However, there is no report in the literature on its cardiovascular effects. AIM OF THE STUDY: The present study was undertaken to assess vasoprotective, anti-atherosclerotic and further examine the anti-dyslipidemic effect of the standardized fraction of Xylocarpus moluccensis (F018) fruits in the mechanical injury and high fat diet (HFD) induced dyslipidemic/ atherosclerosis models. MATERIALS AND METHODS: Guinea pigs were fed 0.08% cholesterol + 15% fat diet for 3 weeks, while ApoE KO mice were fed high fat diet for 18 weeks to induce dyslipidemia and atherosclerosis. A combination of balloon injury and high fat diet (1% cholesterol, 6% peanut oil) for 5 weeks was used to accelerate atherosclerosis in NZW rabbits. F018 was administered once daily by oral route in guinea pigs (10, 25 or 50mg/kg/day for 3 weeks), ApoE KO mice (50mg/kg/day for 6 weeks) and in NZW rabbit (25mg/kg/day for 5 weeks) to monitor its effect on dyslipidemia, vasoreactivity and plaque composition by using standard methodologies. RESULTS: F018 treatment in guinea pigs (25 and 50mg/kg/day), ApoE mice (50mg/kg/day) and rabbits (25mg/kg/day) significantly reduced plasma lipids and improved ACh induced vasorelaxation. Anti-dyslipidemic effect of F018 seems to be due to the modulation of enterohepatic genes involved in the cholesterol absorption and excretion. Moreover, significant improvement in the acetylcholine (ACh) induced vasorelaxation was accompanied with reduced inflammatory burden and enhanced activation of eNOS in ApoE mice aortic tissue. Similarly inflammatory cytokines, immunolabeling of macrophage marker (CD68) and MMP-9 were reduced along with augmentation in vascular smooth muscle cells and collagen type I and III in the mechanically injured iliac artery segment in the rabbits. CONCLUSIONS: Altogether, F018 preserved vasoreactivity, reduced atherosclerotic plaque progression and enhanced plaque stability by reducing lipids, inflammatory cytokines, improving endothelial function and collagen content.


Subject(s)
Atherosclerosis/drug therapy , Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Meliaceae , Plant Extracts/therapeutic use , Animals , Aorta/drug effects , Aorta/physiology , Apolipoproteins E/genetics , Diet, High-Fat , Endothelium, Vascular/drug effects , Fruit , Guinea Pigs , Hypolipidemic Agents/pharmacology , Male , Mice , Mice, Knockout , Plant Extracts/pharmacology , Plaque, Atherosclerotic/drug therapy , Rabbits , Vasodilation/drug effects
15.
Cell Mol Immunol ; 13(6): 745-763, 2016 11.
Article in English | MEDLINE | ID: mdl-26320741

ABSTRACT

In monocytic cells, Toll-like receptor 4 (TLR4)- and TLR2-induced reactive oxygen species (ROS) cause oxidative stress and inflammatory response; however, the mechanism is not well understood. The present study investigated the role of interleukin-1 receptor-associated kinase (IRAK), extracellular signal-regulated kinase (ERK), p67phox and Nox-2 in TLR4- and TLR2-induced ROS generation during interleukin-1 beta (IL-1ß) transcription, processing, and secretion. An IRAK1/4 inhibitor, U0126, PD98059, an NADPH oxidase inhibitor (diphenyleneiodonium (DPI)), and a free radical scavenger (N-acetyl cysteine (NAC))-attenuated TLR4 (lipopolysaccharide (LPS))- and TLR2 (Pam3csk4)-induced ROS generation and IL-1ß production in THP-1 and primary human monocytes. An IRAK1/4 inhibitor and siRNA-attenuated LPS- and Pam3csk4-induced ERK-IRAK1 association and ERK phosphorylation and activity. LPS and Pam3csk4 also induced IRAK1/4-, ERK- and ROS-dependent activation of activator protein-1 (AP-1), IL-1ß transcription, and IL-1ß processing because significant inhibition in AP-1 activity, IL-1ß transcription, Pro- and mature IL-ß expression, and caspase-1 activity was observed with PD98059, U0126, DPI, NAC, an IRAK1/4 inhibitor, tanshinone IIa, and IRAK1 siRNA treatment. IRAK-dependent ERK-p67phox interaction, p67phox translocation, and p67phox-Nox-2 interaction were observed. Nox-2 siRNA significantly reduced secreted IL-1ß, IL-1ß transcript, pro- and mature IL-1ß expression, and caspase-1 activity indicating a role for Nox-2 in LPS- and Pam3csk4-induced IL-1ß production, transcription, and processing. In the present study, we demonstrate that the TLR4- and TLR2-induced IRAK-ERK pathway cross-talks with p67phox-Nox-2 for ROS generation, thus regulating IL-1ß transcription and processing in monocytic cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1beta/genetics , Membrane Glycoproteins/metabolism , Monocytes/metabolism , NADPH Oxidases/metabolism , Phosphoproteins/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Transcription, Genetic , Cell Line , Humans , Interleukin-1beta/metabolism , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Monocytes/drug effects , NADPH Oxidase 2 , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects
16.
Eur J Med Chem ; 110: 1-12, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26807542

ABSTRACT

N-aralkylpyroglutamides of substituted bispidine were prepared and evaluated for their ability to inhibit collagen induced platelet aggregation, both in vivo and in vitro. Some compounds showed high anti-platelet efficacy (in vitro) of which six inhibited both collagen as well as U46619 induced platelet aggregation with concentration dependent anti-platelet efficacy through dual mechanism. In particular, the compound 4j offered significant protection against collagen epinephrine induced pulmonary thromboembolism as well as ferric chloride induced arterial thrombosis, without affecting bleeding tendency in mice. Therefore, the present study suggests that the compound 4j displays a remarkable antithrombotic efficacy much better than aspirin and clopidogrel.


Subject(s)
Blood Platelets/drug effects , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Pulmonary Embolism/prevention & control , Pyrrolidonecarboxylic Acid/therapeutic use , Thrombosis/prevention & control , Animals , Blood Coagulation/drug effects , Blood Platelets/pathology , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Hemorrhage/chemically induced , Humans , Mice , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/chemistry , Pulmonary Embolism/blood , Pulmonary Embolism/pathology , Pyrrolidonecarboxylic Acid/adverse effects , Pyrrolidonecarboxylic Acid/chemical synthesis , Pyrrolidonecarboxylic Acid/chemistry , Thrombosis/blood , Thrombosis/pathology
17.
Front Pharmacol ; 7: 223, 2016.
Article in English | MEDLINE | ID: mdl-27504095

ABSTRACT

The TLR/IL-1R pathway is a critical signaling module that is misregulated in pathologies like inflammation and cancer. Extracts from turmeric (Curcuma longa L.) enriched in curcumin and carbonyls like turmerones have been shown to exert potent anti-inflammatory effects. The present study evaluated the anti-inflammatory activity, cytotoxic effect and the underlying mechanism of a novel chemically modified, non-carbonyl compound enriched Curcuma longa L. (C. longa) extract (CMCE). CMCE (1 or 10 µg/mL; 14 h) significantly decreased LPS (50-100 ng/mL) induced TNF-α and IL-1ß production in THP-1 cells, human, and mouse whole blood as measured by ELISA. LPS-induced IRAK1, MAPK activation, TLR4 expression, TLR4-MyD88 interaction, and IκBα degradation were significantly reduced in CMCE pre-treated THP-1 cells as assessed by Western blotting. CMCE (30, 100, and 300 mg/kg; 10 days p.o.) pre-treated and LPS (10 mg/kg) challenged Swiss mice exhibited attenuated plasma TNF-α, IL-1ß, nitrite, aortic iNOS expression, and vascular dysfunction. In a PI permeability assay, cell lines derived from acute myeloid leukemia were most sensitive to the cytotoxic effects of CMCE. Analysis of Sub-G1 phase, Annexin V-PI positivity, loss of mitochondrial membrane potential, increased caspase-3, and PARP-1 activation confirmed CMCE induced apoptosis in HL-60 cells. IRAK inhibition also sensitized HL-60 cells to CMCE induced cytotoxicity. The present study defines the mechanism underlying the action of CMCE and suggests a therapeutic potential for its use in sepsis and leukemia.

18.
Cell Death Dis ; 7(9): e2348, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27584786

ABSTRACT

Neutrophils play an indispensable role in killing of invading pathogens by enhancing reactive oxygen species (ROS) and NO generation, and subsequently undergoing apoptosis. Unlike ROS/NOX2, role of NO/NOS still remains undefined in the apoptosis of neutrophils (PMNs) and the present study attempts to decipher the importance of NO/NOS in the neutrophil apoptosis. Prolonged treatment of human PMNs or mice bone marrow derived neutrophils (BMDN) with NO led to enhanced ROS generation, caspase-8/caspase-3 cleavage, reduced mitochondrial membrane potential and finally cellular apoptosis. NO-induced ROS generation led to caspase-8 deglutathionylation and activation, which subsequently activated mitochondrial death pathway via BID (Bcl-2 family protein) cleavage. NO-mediated augmentation of caspase-8 and BID cleavage was significantly prevented in BMDN from neutrophil cytosolic factor-1 (NCF-1) knockout (KO) mice, implying the involvement of NOX2 in NO-induced apoptosis of PMNs. Furthermore, ROS, NO generation and inducible nitric oxide synthase (iNOS) expression were enhanced in a time-dependent manner in human PMNs and mice BMDN undergoing spontaneous apoptosis. Pharmacological and genetic ablation of iNOS in human PMNs and mice BMDN significantly reduced the levels of apoptosis. Impaired apoptosis of BMDN from iNOS KO mice was due to reduced caspase-8 activity which subsequently prevented caspase-3 and -9 activation. Altogether, our results suggest a crucial role of NO/iNOS in neutrophil apoptosis via enhanced ROS generation and caspase-8 mediated activation of mitochondrial death pathway.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Neutrophils/cytology , Neutrophils/enzymology , Nitric Oxide/pharmacology , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Enzyme Activation/drug effects , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , NADPH Oxidases/metabolism , Neutrophils/drug effects , Nitric Oxide Synthase Type II/metabolism , Signal Transduction/drug effects
19.
Biochim Biophys Acta ; 1640(1): 77-84, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12676357

ABSTRACT

Mixed lineage kinases (MLKs) belong to the family of mitogen activated protein kinase kinase kinase (MAPKKK) and cause neuronal cell death mediated through c-Jun, N-terminal kinase (JNK) pathway. Recently, genetic studies in Drosophila revealed the presence of an MLK termed slipper (slpr). However, its biochemical features like physiological substrate, role in different MAPK pathways and developmental and tissue-specific expression pattern were not reported. Here, we report cDNA cloning, expression analysis and biochemical characterization of a Drosophila mixed lineage kinase (dMLK) that is also known as slipper. The protein structure analysis of dMLK/slipper revealed, in addition to the conserved domains, a stretch of glutamine in the amino terminus and an asparagine-threonine stretch at the carboxy-terminus. In situ hybridization and reverse transcriptase polymerase chain reaction (RT-PCR) analysis revealed that dMLK is expressed in early embryonic stages, adult brain and thorax. Ectopic expression of dMLK either in Drosophila S2 or in mammalian HEK293 cells leads to activation of JNK, p38 and extracellular signal regulated kinase (ERK) pathways. Further, dMLK directly phosphorylates Hep, dMKK4 and also their mammalian counterparts, MKK7 and SEK1, in an in vitro kinase assay. Taken together, our results provide for the first time a comprehensive expression profile and new biochemical insight of dMLK/slipper.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/enzymology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Signal Transduction , Animals , Cloning, Molecular , Drosophila/embryology , Drosophila Proteins/biosynthesis , Enzyme Activation , In Situ Hybridization , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinases/biosynthesis , Mitogen-Activated Protein Kinases/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
20.
Free Radic Biol Med ; 86: 1-15, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25881549

ABSTRACT

Posttranslational modifications (PTMs) of cytoskeleton proteins due to oxidative stress associated with several pathological conditions often lead to alterations in cell function. The current study evaluates the effect of nitric oxide (DETA-NO)-induced oxidative stress-related S-glutathionylation of cytoskeleton proteins in human PMNs. By using in vitro and genetic approaches, we showed that S-glutathionylation of L-plastin (LPL) and ß-actin promotes reduced chemotaxis, polarization, bactericidal activity, and phagocytosis. We identified Cys-206, Cys-283, and Cys-460as S-thiolated residues in the ß-actin-binding domain of LPL, where cys-460 had the maximum score. Site-directed mutagenesis of LPL Cys-460 further confirmed the role in the redox regulation of LPL. S-Thiolation diminished binding as well as the bundling activity of LPL. The presence of S-thiolated LPL was detected in neutrophils from both diabetic patients and db/db mice with impaired PMN functions. Thus, enhanced nitroxidative stress may results in LPL S-glutathionylation leading to impaired chemotaxis, polarization, and bactericidal activity of human PMNs, providing a mechanistic basis for their impaired functions in diabetes mellitus.


Subject(s)
Actins/metabolism , Microfilament Proteins/metabolism , Neutrophils/physiology , Adult , Amino Acid Sequence , Animals , Case-Control Studies , Cell Polarity , Chemotaxis , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Female , Glutathione/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Obese , Middle Aged , Molecular Sequence Data , Nitric Oxide/metabolism , Oxidative Stress , Protein Binding , Protein Processing, Post-Translational , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL