Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Nutr Food Res ; 67(14): e2200716, 2023 07.
Article in English | MEDLINE | ID: mdl-37150886

ABSTRACT

SCOPE: A prospective study of 34492 participants shows an inverse association between (+)-catechin intake and coronary heart disease. The effects of (+)-catechin on atherosclerosis and associated risk factors are poorly understood and are investigated. METHODS AND RESULTS: (+)-Catechin attenuates reactive oxygen species production in human macrophages, endothelial cells and vascular smooth muscle cells, chemokine-driven monocytic migration, and proliferation of human macrophages and their expression of several pro-atherogenic genes. (+)-Catechin also improves oxidized LDL-mediated mitochondrial membrane depolarization in endothelial cells and attenuates growth factor-induced smooth muscle cell migration. In C57BL/6J mice fed high fat diet (HFD) for 3 weeks, (+)-catechin attenuates plasma levels of triacylglycerol and interleukin (IL)-1ß and IL-2, produces anti-atherogenic changes in liver gene expression, and reduces levels of white blood cells, myeloid-derived suppressor cells, Lin- Sca+ c-Kit+ cells, and common lymphoid progenitor cells within the bone marrow. In LDL receptor deficient mice fed HFD for 12 weeks, (+)-catechin attenuates atherosclerotic plaque burden and inflammation with reduced macrophage content and increased markers of plaque stability; smooth muscle cell and collagen content. CONCLUSION: This study provides novel, detailed insights into the cardio-protective actions of (+)-catechin together with underlying molecular mechanisms and supports further assessments of its beneficial effects in human trials.


Subject(s)
Atherosclerosis , Catechin , Plaque, Atherosclerotic , Humans , Mice , Animals , Plaque, Atherosclerotic/metabolism , Catechin/pharmacology , Catechin/metabolism , Endothelial Cells/metabolism , Mice, Inbred C57BL , Prospective Studies , Mice, Knockout , Atherosclerosis/metabolism , Inflammation/metabolism , Receptors, LDL/metabolism , Risk Factors
2.
Int J Mol Sci ; 24(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36902113

ABSTRACT

Aging and metabolic syndrome are associated with neurodegenerative pathologies including Alzheimer's disease (AD) and there is growing interest in the prophylactic potential of probiotic bacteria in this area. In this study, we assessed the neuroprotective potential of the Lab4P probiotic consortium in both age and metabolically challenged 3xTg-AD mice and in human SH-SY5Y cell culture models of neurodegeneration. In mice, supplementation prevented disease-associated deteriorations in novel object recognition, hippocampal neurone spine density (particularly thin spines) and mRNA expression in hippocampal tissue implying an anti-inflammatory impact of the probiotic, more notably in the metabolically challenged setting. In differentiated human SH-SY5Y neurones challenged with ß-Amyloid, probiotic metabolites elicited a neuroprotective capability. Taken together, the results highlight Lab4P as a potential neuroprotective agent and provide compelling support for additional studies in animal models of other neurodegenerative conditions and human studies.


Subject(s)
Alzheimer Disease , Neuroblastoma , Mice , Humans , Animals , Alzheimer Disease/metabolism , tau Proteins/metabolism , Mice, Transgenic , Neuroblastoma/pathology , Amyloid beta-Peptides/metabolism , Cell Line , Cognition , Disease Models, Animal
3.
Int J Mol Sci ; 24(4)2023 Feb 11.
Article in English | MEDLINE | ID: mdl-36835055

ABSTRACT

Probiotic bacteria have many protective effects against inflammatory disorders, though the mechanisms underlying their actions are poorly understood. The Lab4b consortium of probiotics contains four strains of lactic acid bacteria and bifidobacteria that are reflective of the gut of newborn babies and infants. The effect of Lab4b on atherosclerosis, an inflammatory disorder of the vasculature, has not yet been determined and was investigated on key processes associated with this disease in human monocytes/macrophages and vascular smooth muscle cells in vitro. The Lab4b conditioned medium (CM) attenuated chemokine-driven monocytic migration, monocyte/macrophage proliferation, uptake of modified LDL and macropinocytosis in macrophages together with the proliferation and platelet-derived growth factor-induced migration of vascular smooth muscle cells. The Lab4b CM also induced phagocytosis in macrophages and cholesterol efflux from macrophage-derived foam cells. The effect of Lab4b CM on macrophage foam cell formation was associated with a decrease in the expression of several key genes implicated in the uptake of modified LDL and induced expression of those involved in cholesterol efflux. These studies reveal, for the first time, several anti-atherogenic actions of Lab4b and strongly implicate further studies in mouse models of the disease in vivo and in clinical trials.


Subject(s)
Atherosclerosis , Probiotics , Animals , Mice , Infant, Newborn , Humans , Macrophages/metabolism , Foam Cells/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Lipoproteins, LDL/metabolism
4.
Front Neurosci ; 16: 843105, 2022.
Article in English | MEDLINE | ID: mdl-35685773

ABSTRACT

Brain degenerative disorders such as Alzheimer's disease (AD) can be exacerbated by aberrant metabolism. Supplementation with probiotic bacteria is emerging as a promising preventative strategy for both neurodegeneration and metabolic syndrome. In this study, we assess the impact of the Lab4b probiotic consortium on (i) cognitive and pathological markers of AD progression and (ii) metabolic status in 3xTg-AD mice subjected to metabolic challenge with a high fat diet. The group receiving the probiotic performed better in the novel object recognition test and displayed higher hippocampal neuronal spine density than the control group at the end of the 12 weeks intervention period. These changes were accompanied by differences in localised (brain) and systemic anti-inflammatory responses that favoured the Probiotic group together with the prevention of diet induced weight gain and hypercholesterolaemia and the modulation of liver function. Compositional differences between the faecal microbiotas of the study groups included a lower Firmicutes:Bacteroidetes ratio and less numbers of viable yeast in the Probiotic group compared to the Control. The results illustrate the potential of the Lab4b probiotic as a neuroprotective agent and encourage further studies with human participants.

5.
Front Nutr ; 8: 778289, 2021.
Article in English | MEDLINE | ID: mdl-34901123

ABSTRACT

The anti-inflammatory and cholesterol lowering capabilities of probiotic bacteria highlight them as potential prophylactics against chronic inflammatory diseases, particularly cardiovascular disease. Previous studies in silico, in vitro, and in vivo suggest that the Lab4 probiotic consortium may harbour such capabilities and in the current study, we assessed plasma levels of cytokines/chemokines, short chain fatty acids and lipids and faecal levels of bile acids in a subpopulation of healthy Wistar rats included in 90-day repeat dose oral toxicity study. In the rats receiving Lab4, circulating levels of pro-inflammatory interleukin-6, tumour necrosis factor-α and keratinocyte chemoattractant/growth regulated oncogene were significantly lower compared to the control group demonstrating a systemic anti-inflammatory effect. These changes occurred alongside significant reductions in plasma low density lipoprotein cholesterol and increases in faecal bile acid excretion implying the ability to lower circulating cholesterol via the deconjugation of intestinal bile acids. Correlative analysis identified significant associations between plasma tumour necrosis factor-α and the plasma total cholesterol:high density lipoprotein cholesterol ratio and faecal levels of bifidobacteria in the Lab4 rats. Together, these data highlight Lab4 supplementation as a holistic approach to CVD prevention and encourages further studies in humans.

6.
Mol Nutr Food Res ; 65(17): e2100214, 2021 09.
Article in English | MEDLINE | ID: mdl-34216185

ABSTRACT

SCOPE: Previous studies show that Lab4 probiotic consortium plus Lactobacillus plantarum CUL66 (Lab4P) reduces diet-induced weight gain and plasma cholesterol levels in C57BL/6J mice fed a high fat diet (HFD). The effect of Lab4P on atherosclerosis is not known and is therefore investigated. METHODS AND RESULTS: Atherosclerosis-associated parameters are analyzed in LDL receptor deficient mice fed HFD for 12 weeks alone or supplemented with Lab4P. Lab4P increases plasma HDL and triglyceride levels and decreases LDL/VLDL levels. Lab4P also reduces plaque burden and content of lipids and macrophages, indicative of dampened inflammation, and increases smooth muscle cell content, a marker of plaque stabilization. Atherosclerosis arrays show that Lab4P alters the liver expression of 19 key disease-associated genes. Lab4P also decreases the frequency of macrophages and T-cells in the bone marrow. In vitro assays using conditioned media from probiotic bacteria demonstrates attenuation of several atherosclerosis-associated processes in vitro such as chemokine-driven monocytic migration, proliferation of monocytes and macrophages, foam cell formation and associated changes in expression of key genes, and proliferation and migration of vascular smooth muscle cells. CONCLUSION: This study provides new insights into the anti-atherogenic actions of Lab4P together with the underlying mechanisms and supports further assessments in human trials.


Subject(s)
Atherosclerosis/therapy , Liver/physiology , Plaque, Atherosclerotic/therapy , Probiotics/pharmacology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells , Cholesterol/blood , Culture Media, Conditioned/pharmacology , Diet, High-Fat/adverse effects , Gene Expression Regulation , Lactobacillus plantarum , Lipids/blood , Male , Mice, Mutant Strains , Organ Size , Plaque, Atherosclerotic/pathology , Receptors, LDL/genetics , Spleen/growth & development
7.
Food Funct ; 12(8): 3657-3671, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33900312

ABSTRACT

Atherosclerosis, an inflammatory disorder of the vasculature and the underlying cause of cardiovascular disease, is responsible for one in three global deaths. Consumption of active food ingredients such as omega-3 polyunsaturated fatty acids, flavanols and phytosterols has many beneficial effects on cardiovascular disease. However, their combined actions on the risk factors for atherosclerosis remains poorly understood. We have previously shown that a formulation containing each of these active components at physiologically relevant doses modulated several monocyte/macrophage processes associated with atherosclerosis in vitro, including inhibition of cytokine-induced pro-inflammatory gene expression, chemokine-driven monocyte migration, expression of M1 phenotype markers, and promotion of cholesterol efflux. The objectives of the present study were to investigate whether the protective actions of the formulation extended in vivo and to delineate the potential underlying mechanisms. The formulation produced several favourable changes, including higher plasma levels of HDL and reduced levels of macrophages and myeloid-derived suppressor cells in the bone marrow. The mRNA expression of liver-X-receptor-α, peroxisome proliferator-activated receptor-γ and superoxide dismutase-1 was induced in the liver and that of interferon-γ and the chemokine (C-X-C motif) ligand 1 decreased, thereby suggesting the potential mechanisms for many beneficial effects. Other changes were also observed such as increased plasma levels of triglycerides and lipid peroxidation that may reflect potential activation of brown fat. This study provides new insights into the protective actions and the potential underlying mechanisms of the formulation in vivo, particularly in relation to risk factors together with changes in systemic inflammation and hepatic lipid alterations associated with atherosclerosis and metabolic syndrome, and supports further assessments in human trials.


Subject(s)
Cardiotonic Agents/pharmacology , Coronary Artery Disease/prevention & control , Animals , Cardiotonic Agents/administration & dosage , Diet, High-Fat , Disease Models, Animal , Fatty Acids, Omega-3/administration & dosage , Flavanones/administration & dosage , Functional Food , Gene Expression , Male , Mice , Mice, Inbred C57BL , Phytosterols/administration & dosage , Risk Factors
8.
Gut Microbes ; 13(1): 1-9, 2021.
Article in English | MEDLINE | ID: mdl-33764850

ABSTRACT

Gut microbiome manipulation to alter the gut-lung axis may potentially protect humans against respiratory infections, and clinical trials of probiotics show promise in this regard in healthy adults and children. However, comparable studies are lacking in overweight/obese people, who have increased risks in particular of viral upper respiratory tract infections (URTI). This Addendum further analyses our recent placebo-controlled trial of probiotics in overweight/obese people (focused initially on weight loss) to investigate the impact of probiotics upon the occurrence of URTI symptoms. As well as undergoing loss of weight and improvement in certain metabolic parameters, study participants taking probiotics experienced a 27% reduction in URTI symptoms versus control, with those ≥45 years or BMI ≥30 kg/m2 experiencing greater reductions. This symptom reduction is apparent within 2 weeks of probiotic use. Gut microbiome diversity remained stable throughout the study in probiotic-treated participants. Our data provide support for further trials to assess the potential role of probiotics in preventing viral URTI (and possibly also COVID-19), particularly in overweight/obese people.


Subject(s)
Obesity/complications , Overweight/complications , Probiotics/therapeutic use , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/therapy , Adult , Aged , Double-Blind Method , Gastrointestinal Microbiome , Humans , Middle Aged , Pandemics , Self Report
10.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2538-2550, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31202985

ABSTRACT

Atherosclerosis and its complications are responsible for one in three global deaths. Nutraceuticals show promise in the prevention and treatment of atherosclerosis but require an indepth understanding of the mechanisms underlying their actions. A previous study showed that the omega-6 fatty acid, dihomo-γ-linolenic acid (DGLA), attenuated atherosclerosis in the apolipoprotein E deficient mouse model system. However, the mechanisms underlying such protective effects of DGLA are poorly understood and were therefore investigated. We show that DGLA attenuates chemokine-driven monocytic migration together with foam cell formation and the expression of key pro-atherogenic genes induced by three pro-inflammatory cytokines in human macrophages. The effect of DGLA on interferon-γ signaling was mediated via inhibition of signal transducer and activator of transcription-1 phosphorylation on serine 727. In relation to anti-foam cell action, DGLA inhibits modified LDL uptake by both macropinocytosis and receptor-mediated endocytosis, the latter by reduction in expression of two key scavenger receptors (SR-A and CD36), and stimulates cholesterol efflux from foam cells. DGLA also improves macrophage mitochondrial bioenergetic profile by decreasing proton leak. Gamma-linolenic acid and prostaglandin E1, upstream precursor and key metabolite respectively of DGLA, also acted in an anti-atherogenic manner. The actions of DGLA extended to other key atherosclerosis-associated cell types with attenuation of endothelial cell proliferation and migration of smooth muscle cells in response to platelet-derived growth factor. This study provides novel insights into the molecular mechanisms underlying the anti-atherogenic actions of DGLA and supports further assessments on its protective effects on plaque regression in vivo and in human trials.


Subject(s)
8,11,14-Eicosatrienoic Acid/pharmacology , Gene Expression Regulation/drug effects , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cytokines/metabolism , Foam Cells/cytology , Foam Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Monocytes/cytology
11.
Sci Rep ; 6: 34368, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27687241

ABSTRACT

The anti-atherogenic cytokine TGF-ß inhibits macrophage foam cell formation by suppressing the expression of key genes implicated in the uptake of modified lipoproteins. We have previously shown a critical role for p38 MAPK and JNK in the TGF-ß-mediated regulation of apolipoprotein E expression in human monocytes. However, the roles of these two MAPK pathways in the control of expression of key genes involved in the uptake of modified lipoproteins in human macrophages is poorly understood and formed the focus of this study. TGF-ß activated both p38 MAPK and JNK, and knockdown of p38 MAPK or c-Jun, a key downstream target of JNK action, demonstrated their requirement in the TGF-ß-inhibited expression of several key genes implicated in macrophage lipoprotein uptake. The potential role of c-Jun and specific co-activators in the action of TGF-ß was investigated further by studies on the lipoprotein lipase gene. c-Jun did not directly interact with the minimal promoter region containing the TGF-ß response elements and a combination of transient transfection and knock down assays revealed an important role for SRC-1. These studies provide novel insights into the mechanisms underlying the TGF-ß-mediated inhibition of macrophage gene expression associated with the control of cholesterol homeostasis.

12.
PLoS One ; 11(3): e0151057, 2016.
Article in English | MEDLINE | ID: mdl-26950833

ABSTRACT

INTRODUCTION: Atherosclerosis is the underlying cause of cardiovascular disease that leads to more global mortalities each year than any other ailment. Consumption of active food ingredients such as phytosterols, omega-3 polyunsaturated fatty acids and flavanols are known to impart beneficial effects on cardiovascular disease although the combined actions of such agents in atherosclerosis is poorly understood. The aim of this study was to screen a nutritional supplement containing each of these active components for its anti-atherosclerotic effect on macrophages in vitro. RESULTS: The supplement attenuated the expression of intercellular adhesion molecule-1 and macrophage chemoattractant protein-1 in human and murine macrophages at physiologically relevant doses. The migratory capacity of human monocytes was also hindered, possibly mediated by eicosapentaenoic acid and catechin, while the ability of foam cells to efflux cholesterol was improved. The polarisation of murine macrophages towards a pro-inflammatory phenotype was also attenuated by the supplement. CONCLUSION: The formulation was able to hinder multiple key steps of atherosclerosis development in vitro by inhibiting monocyte recruitment, foam cell formation and macrophage polarisation towards an inflammatory phenotype. This is the first time a combination these ingredients has been shown to elicit such effects and supports its further study in preclinical in vivo models.


Subject(s)
Atherosclerosis/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Flavonols/pharmacology , Phytosterols/pharmacology , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biological Transport/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chemokine CCL2/genetics , Cholesterol/metabolism , Drug Interactions , Gene Expression Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Monocytes/cytology , Monocytes/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
J Cell Physiol ; 2015 May 07.
Article in English | MEDLINE | ID: mdl-25953328

ABSTRACT

The transcription of the ATP-binding cassette transporter A1 (ABCA1) gene, which plays a key anti-atherogenic role, is known to be induced by agonists of liver X receptors (LXRs). LXRs form obligate heterodimers with retinoid X receptors (RXRs) and interact with their recognition sequences in the regulatory regions of key genes implicated in the control of cholesterol, fatty acid and glucose homeostasis. We have previously shown a novel role for c-Jun N-terminal kinase (JNK) and phosphoinositide 3-kinase (PI3K) in the LXRs-mediated induction of macrophage gene expression. Protein kinase C (PKC) is often found to regulate the action of nuclear receptors and cross talk between this kinase family and JNK and/or PI3K has been shown in several settings. We have therefore investigated a potential role for PKC in the action of LXR/RXR agonist 22-(R)-hydroxycholesterol (22-(R)-HC)/9-cis-retinoic acid (9cRA) in THP-1 macrophages, including the induction of ABCA1 expression. The pan PKC inhibitor bisindoylmaleimide was found to attenuate the induction of ABCA1 protein expression, the activation of the JNK signaling pathway and the stimulation of activator protein-1 (AP-1) DNA binding activity in macrophages treated with 22-(R)-HC and 9cRA. The role of PKC in the action of these ligands was confirmed further by the use of more isotype-specific inhibitors. These studies therefore reveal a potentially important role for PKC in the action of 22-(R)-HC and 9cRA in human macrophages. This article is protected by copyright. All rights reserved.

14.
J Cell Biochem ; 116(9): 2032-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25752819

ABSTRACT

The transcription of the ATP-binding cassette transporter A1 (ABCA1) gene, which plays a key anti-atherogenic role, is known to be induced by agonists of liver X receptors (LXRs). LXRs form obligate heterodimers with retinoid X receptors (RXRs) and interact with their recognition sequences in the regulatory regions of key genes implicated in the control of cholesterol, fatty acid and glucose homeostasis. We have previously shown a novel role for c-Jun N-terminal kinase (JNK) and phosphoinositide 3-kinase (PI3K) in the LXRs-mediated induction of macrophage gene expression. Protein kinase C (PKC) is often found to regulate the action of nuclear receptors and cross talk between this kinase family and JNK and/or PI3K has been shown in several settings. We have, therefore, investigated a potential role for PKC in the action of LXR/RXR agonist 22-(R)-hydroxycholesterol (22-(R)-HC)/9-cis-retinoic acid (9cRA) in THP-1 macrophages, including the induction of ABCA1 expression. The pan PKC inhibitor bisindoylmaleimide was found to attenuate the induction of ABCA1 protein expression, the activation of the JNK signaling pathway and the stimulation of activator protein-1 (AP-1) DNA binding activity in macrophages treated with 22-(R)-HC and 9cRA. The role of PKC in the action of these ligands was confirmed further by the use of more isotype-specific inhibitors. These studies, therefore, reveal a potentially important role for PKC in the action of 22-(R)-HC and 9cRA in human macrophages.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Hydroxycholesterols/pharmacology , Macrophages/drug effects , Protein Kinase C/metabolism , Tretinoin/pharmacology , ATP Binding Cassette Transporter 1/antagonists & inhibitors , Alitretinoin , Cell Line , Gene Expression Regulation/drug effects , Humans , Indoles/pharmacology , Liver X Receptors , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Maleimides/pharmacology , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/metabolism
15.
Lipids ; 50(3): 253-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25663263

ABSTRACT

The transformation of macrophages into lipid-loaded foam cells is a critical early event in the pathogenesis of atherosclerosis. Both receptor-mediated uptake of modified LDL, mediated primarily by scavenger receptors-A (SR-A) and CD36 along with other proteins such as lipoprotein lipase (LPL), and macropinocytosis contribute to macrophage foam cell formation. The signaling pathways that are involved in the control of foam cell formation are not fully understood. In this study, we have investigated the role of phosphoinositide 3-kinase (PI3K) in relation to foam cell formation in human macrophages. The pan PI3K inhibitor LY294002 attenuated the uptake of modified LDL and macropinocytosis, as measured by Lucifer Yellow uptake, by human macrophages. In addition, the expression of SR-A, CD36 and LPL was attenuated by LY294002. The use of isoform-selective PI3K inhibitors showed that PI3K-ß, -γ and -δ were all required for the expression of SR-A and CD36 whereas only PI3K-γ was necessary in the case of LPL. These studies reveal a pivotal role of PI3K in the control of macrophage foam cell formation and provide further evidence for their potential as therapeutic target against atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Foam Cells/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Atherosclerosis/metabolism , Chromones/pharmacology , Gene Expression/drug effects , Humans , Macrophages/drug effects , Macrophages/pathology , Morpholines/pharmacology
16.
Cytokine ; 64(1): 357-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23791479

ABSTRACT

A key event during the formation of lipid-rich foam cells during the progression of atherosclerosis is the uptake of modified low-density lipoproteins (LDL) by macrophages in response to atherogenic mediators in the arterial intima. In addition to scavenger receptor-dependent uptake of LDL, macropinocytosis is known to facilitate the uptake of LDL through the constitutive and passive internalization of large quantities of extracellular solute. In this study we confirm the ability of macropinocytosis to facilitate the uptake of modified LDL by human macrophages and show its modulation by TGF-ß, IFN-γ, IL-17A and IL-33. Furthermore we show that the TGF-ß-mediated inhibition of macropinocytosis is a Smad-2/-3-independent process.


Subject(s)
Atherosclerosis/pathology , Foam Cells/pathology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Pinocytosis/drug effects , Transforming Growth Factor beta/metabolism , Atherosclerosis/immunology , Biological Transport/drug effects , Cell Differentiation , Cells, Cultured , Cytochalasin D/pharmacology , Foam Cells/immunology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Humans , Inflammation/immunology , Interleukin-33 , Lipoproteins, LDL/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
17.
Matrix Biol ; 31(7-8): 373-9, 2012.
Article in English | MEDLINE | ID: mdl-23123404

ABSTRACT

The ubiquitous mammalian extracellular matrix glycosaminoglycan hyaluronan (HA) plays a pivotal role in the regulation of cell phenotype in fibrosis and scarring. Transforming growth factor-beta 1 (TGF-ß1) and interleukin-1 beta (IL-1ß) up-regulate hyaluronan synthase (HAS) 1 and HAS2 in dermal fibroblasts and renal proximal tubular epithelial cells, and subsequent HA synthesis regulates cell phenotype. In the present study, we investigated the mechanism of HAS1 transcriptional up-regulation in response to these cytokines. We used 5'-rapid amplification of cDNA ends analysis to identify the 5' end of HAS1 transcripts, resulting in an increase of 26 nucleotides to the HAS1 exon 1 sequence of reference sequence NM_001523. Constitutive luciferase activity of upstream DNA sequences was shown in luciferase reporter assays, but our reporter vector signals were refractory to the addition of TGF-ß1 and IL-1ß. Using siRNAs to knockdown transcription factor mRNAs, we showed that TGF-ß1 up-regulation of HAS1 transcription was mediated via Smad3 but not Smad2, while HAS1 induction by IL-1ï€ ß was Sp3, not Sp1, dependent.


Subject(s)
Extracellular Matrix Proteins/genetics , Gene Expression Regulation/genetics , Glucuronosyltransferase/genetics , Promoter Regions, Genetic/genetics , Smad3 Protein/metabolism , Sp3 Transcription Factor/metabolism , Base Sequence , DNA Primers/genetics , DNA, Complementary/genetics , Electrophoresis, Agar Gel , Gene Expression Regulation/physiology , Humans , Hyaluronan Synthases , Interleukin-1beta/metabolism , Luciferases , Molecular Sequence Data , Nucleic Acid Amplification Techniques , RNA Interference , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
18.
Biochim Biophys Acta ; 1822(10): 1608-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705205

ABSTRACT

The anti-atherogenic cytokine, TGF-ß, plays a key role during macrophage foam cell formation by modulating the expression of key genes involved in the control of cholesterol homeostasis. Unfortunately, the molecular mechanisms underlying these actions of TGF-ß remain poorly understood. In this study we examine the effect of TGF-ß on macrophage cholesterol homeostasis and delineate the role of Smads-2 and -3 during this process. Western blot analysis showed that TGF-ß induces a rapid phosphorylation-dependent activation of Smad-2 and -3 in THP-1 and primary human monocyte-derived macrophages. Small interfering RNA-mediated knockdown of Smad-2/3 expression showed that the TGF-ß-mediated regulation of key genes implicated in the uptake of modified low density lipoproteins and the efflux of cholesterol from foam cells was Smad-dependent. Additionally, through the use of virally delivered Smad-2 and/or Smad-3 short hairpin RNA, we demonstrate that TGF-ß inhibits the uptake of modified LDL by macrophages through a Smad-dependent mechanism and that the TGF-ß-mediated regulation of CD36, lipoprotein lipase and scavenger receptor-A gene expression was dependent on Smad-2. These studies reveal a crucial role for Smad signaling, particularly Smad-2, in the inhibition of foam cell formation by TGF-ß through the regulation of expression of key genes involved in the control of macrophage cholesterol homeostasis.


Subject(s)
Lipoproteins, LDL/metabolism , Macrophages/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cells, Cultured , Cholesterol/genetics , Cholesterol/metabolism , Foam Cells/metabolism , Gene Expression , Homeostasis , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Lipoproteins, LDL/genetics , Phosphorylation , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics
19.
Curr Atheroscler Rep ; 14(3): 284-93, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22419222

ABSTRACT

Liver X receptors (LXRs) belong to the nuclear receptor superfamily of ligand-dependent transcription factors. LXRs are activated by oxysterols, metabolites of cholesterol, and therefore act as intracellular sensors of this lipid. There are two LXR genes (α and ß) that display distinct tissue/cell expression profiles. LXRs interact with regulatory sequences in target genes as heterodimers with retinoid X receptor. Such direct targets of LXR actions include important genes implicated in the control of lipid homeostasis, particularly reverse cholesterol transport. In addition, LXRs attenuate the transcription of genes associated with the inflammatory response indirectly by transrepression. In this review, we describe recent evidence that both highlights the key roles of LXRs in atherosclerosis and inflammation and provides novel insights into the mechanisms underlying their actions. In addition, we discuss the major limitations of LXRs as therapeutic targets for the treatment of atherosclerosis and how these are being addressed.


Subject(s)
Atherosclerosis/metabolism , Inflammation/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Orphan Nuclear Receptors/physiology , Animals , Humans , Liver X Receptors
20.
Lipids ; 46(11): 1053-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21822944

ABSTRACT

There is evidence that long chain n-3 PUFA (such as from fish oils) provide atheroprotection through, in part, changes in macrophage function although it has not been fully determined whether these n-3 PUFA target cellular mechanisms that control macrophage foam cell formation. Therefore, we investigated whether the n-3 PUFA, EPA and DHA, modulate modified low-density lipoprotein (LDL) uptake by human macrophages. The uptake of fluorophore labeled acetylated LDL (AcLDL) and/or oxidized LDL (OxLDL) by THP-1 macrophages and primary human monocyte-derived macrophages were measured by flow cytometry following co-incubation with EPA or DHA in vitro. DHA inhibited both AcLDL and OxLDL uptake in human macrophages whilst EPA reduced AcLDL and increased OxLDL uptake. These effects were only partly explained by changes in the mRNA and protein expression of key scavenger receptors, such as CD36 and scavenger receptor-A, in these cells suggesting the involvement of a scavenger receptor-independent mechanism. EPA and DHA inhibited macropinocytosis, as measured by Lucifer Yellow uptake, in human macrophages and attenuated the expression of Syndecan-4, which has been implicated in the uptake of other modified forms of LDL. EPA and DHA reduced modified LDL uptake by human macrophages through a mechanism that is in part scavenger receptor-independent and may involve inhibition of macropinocytosis and Syndecan-4 expression. This suggests that both EPA and DHA are capable of regulating macrophage foam cell formation and adds to the evidence describing an atheroprotective role for n-3 PUFA, implicating them as potential therapeutic agents for the treatment of clinical atherosclerosis.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Lipoproteins, LDL/metabolism , Macrophages/physiology , Pinocytosis/drug effects , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Line , Gene Expression , Humans , Macrophages/drug effects , Macrophages/metabolism , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Syndecan-4/genetics , Syndecan-4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...