Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 16: 1299314, 2023.
Article in English | MEDLINE | ID: mdl-38164407

ABSTRACT

Microglia are crucial for brain homeostasis, and dysfunction of these cells is a key driver in most neurodegenerative diseases, including peroxisomal leukodystrophies. In X-linked adrenoleukodystrophy (X-ALD), a neuroinflammatory disorder, very long-chain fatty acid (VLCFA) accumulation due to impaired degradation within peroxisomes results in microglial defects, but the underlying mechanisms remain unclear. Using CRISPR/Cas9 gene editing of key genes in peroxisomal VLCFA breakdown (Abcd1, Abcd2, and Acox1), we recently established easily accessible microglial BV-2 cell models to study the impact of dysfunctional peroxisomal ß-oxidation and revealed a disease-associated microglial-like signature in these cell lines. Transcriptomic analysis suggested consequences on the immune response. To clarify how impaired lipid degradation impacts the immune function of microglia, we here used RNA-sequencing and functional assays related to the immune response to compare wild-type and mutant BV-2 cell lines under basal conditions and upon pro-inflammatory lipopolysaccharide (LPS) activation. A majority of genes encoding proinflammatory cytokines, as well as genes involved in phagocytosis, antigen presentation, and co-stimulation of T lymphocytes, were found differentially overexpressed. The transcriptomic alterations were reflected by altered phagocytic capacity, inflammasome activation, increased release of inflammatory cytokines, including TNF, and upregulated response of T lymphocytes primed by mutant BV-2 cells presenting peptides. Together, the present study shows that peroxisomal ß-oxidation defects resulting in lipid alterations, including VLCFA accumulation, directly reprogram the main cellular functions of microglia. The elucidation of this link between lipid metabolism and the immune response of microglia will help to better understand the pathogenesis of peroxisomal leukodystrophies.

2.
Nutrients ; 14(11)2022 May 25.
Article in English | MEDLINE | ID: mdl-35683996

ABSTRACT

Coronavirus illness (COVID-19) is an infectious pathology generated by intense severe respiratory syndrome coronavirus 2 (SARS-CoV-2). This infectious disease has emerged in 2019. The COVID-19-associated pandemic has considerably affected the way of life and the economy in the world. It is consequently crucial to find solutions allowing remedying or alleviating the effects of this infectious disease. Natural products have been in perpetual application from immemorial time given that they are attested to be efficient towards several illnesses without major side effects. Various studies have shown that plant extracts or purified molecules have a promising inhibiting impact towards coronavirus. In addition, it is substantial to understand the characteristics, susceptibility and impact of diet on patients infected with COVID-19. In this review, we recapitulate the influence of extracts or pure molecules from medicinal plants on COVID-19. We approach the possibilities of plant treatment/co-treatment and feeding applied to COVID-19. We also show coronavirus susceptibility and complications associated with nutrient deficiencies and then discuss the major food groups efficient on COVID-19 pathogenesis. Then, we covered emerging technologies using plant-based SARS-CoV-2 vaccine. We conclude by giving nutrient and plants curative therapy recommendations which are of potential interest in the COVID-19 infection and could pave the way for pharmacological treatments or co-treatments of COVID-19.


Subject(s)
COVID-19 , Antiviral Agents/therapeutic use , COVID-19 Vaccines , Diet , Humans , Incidence , Nutrients , Oxidative Stress , SARS-CoV-2
3.
J Lipid Res ; 62: 100011, 2021.
Article in English | MEDLINE | ID: mdl-33500240

ABSTRACT

Bacterial lipopolysaccharides (LPSs or endotoxins) can bind most proteins of the lipid transfer/LPS-binding protein (LT/LBP) family in host organisms. The LPS-bound LT/LBP proteins then trigger either an LPS-induced proinflammatory cascade or LPS binding to lipoproteins that are involved in endotoxin inactivation and detoxification. Cholesteryl ester transfer protein (CETP) is an LT/LBP member, but its impact on LPS metabolism and sepsis outcome is unclear. Here, we performed fluorescent LPS transfer assays to assess the ability of CETP to bind and transfer LPS. The effects of intravenous (iv) infusion of purified LPS or polymicrobial infection (cecal ligation and puncture [CLP]) were compared in transgenic mice expressing human CETP and wild-type mice naturally having no CETP activity. CETP displayed no LPS transfer activity in vitro, but it tended to reduce biliary excretion of LPS in vivo. The CETP expression in mice was associated with significantly lower basal plasma lipid levels and with higher mortality rates in both models of endotoxemia and sepsis. Furthermore, CETPTg plasma modified cytokine production of macrophages in vitro. In conclusion, despite having no direct LPS binding and transfer property, human CETP worsens sepsis outcomes in mice by altering the protective effects of plasma lipoproteins against endotoxemia, inflammation, and infection.


Subject(s)
Cholesterol Ester Transfer Proteins
4.
J Clin Invest ; 130(11): 5858-5874, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32759503

ABSTRACT

Mitochondria have emerged as key actors of innate and adaptive immunity. Mitophagy has a pivotal role in cell homeostasis, but its contribution to macrophage functions and host defense remains to be delineated. Here, we showed that lipopolysaccharide (LPS) in combination with IFN-γ inhibited PINK1-dependent mitophagy in macrophages through a STAT1-dependent activation of the inflammatory caspases 1 and 11. In addition, we demonstrated that the inhibition of mitophagy triggered classical macrophage activation in a mitochondrial ROS-dependent manner. In a murine model of polymicrobial infection (cecal ligature and puncture), adoptive transfer of Pink1-deficient bone marrow or pharmacological inhibition of mitophagy promoted macrophage activation, which favored bactericidal clearance and led to a better survival rate. Reciprocally, mitochondrial uncouplers that promote mitophagy reversed LPS/IFN-γ-mediated activation of macrophages and led to immunoparalysis with impaired bacterial clearance and lowered survival. In critically ill patients, we showed that mitophagy was inhibited in blood monocytes of patients with sepsis as compared with nonseptic patients. Overall, this work demonstrates that the inhibition of mitophagy is a physiological mechanism that contributes to the activation of myeloid cells and improves the outcome of sepsis.


Subject(s)
Bacteria/immunology , Macrophage Activation , Macrophages, Peritoneal/immunology , Mitophagy/immunology , Sepsis/immunology , Animals , Female , Humans , Interferon-gamma/immunology , Lipopolysaccharides/immunology , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Male , Mice , Protein Kinases/immunology , RAW 264.7 Cells , Sepsis/microbiology , Sepsis/pathology
5.
mBio ; 10(3)2019 06 04.
Article in English | MEDLINE | ID: mdl-31164474

ABSTRACT

Secondary bacterial lung infection by Streptococcus pneumoniae (S. pneumoniae) poses a serious health concern, especially in developing countries. We posit that the emergence of multiantibiotic-resistant strains will jeopardize current treatments in these regions. Deaths arising from secondary infections are more often associated with acute lung injury, a common consequence of hypercytokinemia, than with the infection per se Given that secondary bacterial pneumonia often has a poor prognosis, newer approaches to improve treatment outcomes are urgently needed to reduce the high levels of morbidity and mortality. Using a sequential dual-infection mouse model of secondary bacterial lung infection, we show that host-directed therapy via immunoneutralization of the angiopoietin-like 4 c-isoform (cANGPTL4) reduced pulmonary edema and damage in infected mice. RNA sequencing analysis revealed that anti-cANGPTL4 treatment improved immune and coagulation functions and reduced internal bleeding and edema. Importantly, anti-cANGPTL4 antibody, when used concurrently with either conventional antibiotics or antipneumolysin antibody, prolonged the median survival of mice compared to monotherapy. Anti-cANGPTL4 treatment enhanced immune cell phagocytosis of bacteria while restricting excessive inflammation. This modification of immune responses improved the disease outcomes of secondary pneumococcal pneumonia. Taken together, our study emphasizes that host-directed therapeutic strategies are viable adjuncts to standard antimicrobial treatments.IMPORTANCE Despite extensive global efforts, secondary bacterial pneumonia still represents a major cause of death in developing countries and is an important cause of long-term functional disability arising from lung tissue damage. Newer approaches to improving treatment outcomes are needed to reduce the significant morbidity and mortality caused by infectious diseases. Our study, using an experimental mouse model of secondary S. pneumoniae infection, shows that a multimodal treatment that concurrently targets host and pathogen factors improved lung tissue integrity and extended the median survival time of infected mice. The immunoneutralization of host protein cANGPTL4 reduced the severity of pulmonary edema and damage. We show that host-directed therapeutic strategies as well as neutralizing antibodies against pathogen virulence factors are viable adjuncts to standard antimicrobial treatments such as antibiotics. In view of their different modes of action compared to antibiotics, concurrent immunotherapies using antibodies are potentially efficacious against secondary pneumococcal pneumonia caused by antibiotic-resistant pathogens.


Subject(s)
Angiopoietin-Like Protein 4/antagonists & inhibitors , Antibodies/therapeutic use , Coinfection/therapy , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/therapy , Pulmonary Edema/therapy , Angiopoietin-Like Protein 4/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Coinfection/immunology , Coinfection/microbiology , Disease Models, Animal , Female , Inflammation , Lung/microbiology , Mice , Mice, Inbred BALB C , Pneumonia, Pneumococcal/drug therapy , Pulmonary Edema/immunology , Streptococcus pneumoniae/immunology
6.
Cell Rep ; 21(5): 1160-1168, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29091756

ABSTRACT

Glucagon-like peptide 1 (GLP-1) is a hormone released from enteroendocrine L cells. Although first described as a glucoregulatory incretin hormone, GLP-1 also suppresses inflammation and promotes mucosal integrity. Here, we demonstrate that plasma GLP-1 levels are rapidly increased by lipopolysaccharide (LPS) administration in mice via a Toll-like receptor 4 (TLR4)-dependent mechanism. Experimental manipulation of gut barrier integrity after dextran sodium sulfate treatment, or via ischemia/reperfusion experiments in mice, triggered a rapid rise in circulating GLP-1. This phenomenon was detected prior to measurable changes in inflammatory status and plasma cytokine and LPS levels. In human subjects, LPS administration also induced GLP-1 secretion. Furthermore, GLP-1 levels were rapidly increased following the induction of ischemia in the human intestine. These findings expand traditional concepts of enteroendocrine L cell biology to encompass the sensing of inflammatory stimuli and compromised mucosal integrity, linking glucagon-like peptide secretion to gut inflammation.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Ileum/drug effects , Lipopolysaccharides/toxicity , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Calcium Signaling/drug effects , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate/pharmacology , Enteroendocrine Cells/cytology , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Humans , Ileum/metabolism , Interleukin-6/deficiency , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myristic Acids/blood , Proglucagon/metabolism , Proprotein Convertase 1/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Young Adult
7.
Int J Mol Sci ; 18(10)2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29048364

ABSTRACT

Sepsis causes severe dysregulation of organ functions, via the development of oxidative stress and inflammation. These pathophysiological mechanisms are mimicked in mice injected with bacterial lipopolysaccharide (LPS). Here, protective properties of argan oil against LPS-induced oxidative stress and inflammation are explored in the murine model. Mice received standard chow, supplemented with argan oil (AO) or olive oil (OO) for 25 days, before septic shock was provoked with a single intraperitoneal injection of LPS, 16 hours prior to animal sacrifice. In addition to a rise in oxidative stress and inflammatory markers, injected LPS also caused hepatotoxicity, accompanied by hyperglycemia, hypercholesterolemia and hyperuremia. These LPS-associated toxic effects were blunted by AO pretreatment, as corroborated by normal plasma parameters and cell stress markers (glutathione: GSH) and antioxidant enzymology (catalase, CAT; superoxide dismutase, SOD and glutathione peroxidase, GPx). Hematoxylin-eosin staining revealed that AO can protect against acute liver injury, maintaining a normal status, which is pointed out by absent or reduced LPS-induced hepatic damage markers (i.e., alanine aminotransferase (ALT) and aspartate transaminase (AST)). Our work also indicated that AO displayed anti-inflammatory activity, due to down-regulations of genes encoding pro-inflammatory cytokines Interleukin-6 (IL-6) and Tumor Necrosis Factor-α (TNF-α) and in up-regulations of the expression of anti-inflammatory genes encoding Interleukin-4 (IL-4) and Interleukin-10 (IL-10). OO provided animals with similar, though less extensive, protective changes. Collectively our work adds compelling evidence to the protective mechanisms of AO against LPS-induced liver injury and hence therapeutic potentialities, in regard to the management of human sepsis. Activations of IL-4/Peroxisome Proliferator-Activated Receptors (IL-4/PPARs) signaling and, under LPS, an anti-inflammatory IL-10/Liver X Receptor (IL-10/LXR) route, obviously indicated the high potency and plasticity of the anti-inflammatory properties of argan oil.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Liver Diseases/drug therapy , Liver/drug effects , Olive Oil/pharmacology , Oxidative Stress , Plant Oils/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Dietary Supplements , Lipopolysaccharides/toxicity , Liver/metabolism , Liver Diseases/etiology , Liver Diseases/prevention & control , Mice , Olive Oil/administration & dosage , Olive Oil/therapeutic use , Plant Oils/administration & dosage , Plant Oils/therapeutic use
8.
J Lipid Res ; 58(7): 1399-1416, 2017 07.
Article in English | MEDLINE | ID: mdl-28533304

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, yet the pathogenesis of NAFLD is only partially understood. Here, we investigated the role of the gut bacteria in NAFLD by stimulating the gut bacteria via feeding mice the fermentable dietary fiber, guar gum (GG), and suppressing the gut bacteria via chronic oral administration of antibiotics. GG feeding profoundly altered the gut microbiota composition, in parallel with reduced diet-induced obesity and improved glucose tolerance. Strikingly, despite reducing adipose tissue mass and inflammation, GG enhanced hepatic inflammation and fibrosis, concurrent with markedly elevated plasma and hepatic bile acid levels. Consistent with a role of elevated bile acids in the liver phenotype, treatment of mice with taurocholic acid stimulated hepatic inflammation and fibrosis. In contrast to GG, chronic oral administration of antibiotics effectively suppressed the gut bacteria, decreased portal secondary bile acid levels, and attenuated hepatic inflammation and fibrosis. Neither GG nor antibiotics influenced plasma lipopolysaccharide levels. In conclusion, our data indicate a causal link between changes in gut microbiota and hepatic inflammation and fibrosis in a mouse model of NAFLD, possibly via alterations in bile acids.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Biological Transport , Diet, High-Fat/adverse effects , Fibrosis , Galactans/adverse effects , Gastrointestinal Microbiome/drug effects , Glucose Tolerance Test , Liver/metabolism , Liver/pathology , Male , Mannans/adverse effects , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/pathology , Obesity/chemically induced , Obesity/metabolism , Obesity/microbiology , Plant Gums/adverse effects
9.
Molecules ; 22(1)2017 Jan 07.
Article in English | MEDLINE | ID: mdl-28067864

ABSTRACT

In this study, we aimed to evaluate the antioxidant and anti-inflammatory properties of Opuntia ficus-indica cactus cladode extracts in microglia BV-2 cells. Inflammation associated with microglia activation in neuronal injury can be achieved by LPS exposure. Using four different structurally and biologically well-characterized LPS serotypes, we revealed a structure-related differential effect of LPS on fatty acid ß-oxidation and antioxidant enzymes in peroxisomes: Escherichia coli-LPS decreased ACOX1 activity while Salmonella minnesota-LPS reduced only catalase activity. Different cactus cladode extracts showed an antioxidant effect through microglial catalase activity activation and an anti-inflammatory effect by reducing nitric oxide (NO) LPS-dependent production. These results suggest that cactus extracts may possess a neuroprotective activity through the induction of peroxisomal antioxidant activity and the inhibition of NO production by activated microglial cells.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Catalase/metabolism , Microglia/metabolism , Nitric Oxide/metabolism , Opuntia/chemistry , Peroxisomes/metabolism , Plant Extracts/pharmacology , Animals , Cell Line , Escherichia coli , Fatty Acids/metabolism , Lipopolysaccharides , Mice , Microglia/cytology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Salmonella
10.
Biochim Open ; 1: 51-59, 2015.
Article in English | MEDLINE | ID: mdl-29632829

ABSTRACT

In patients with sepsis, liver metabolism and its capacity to provide other organs with energetic substrates are impaired. This and many other pathophysiological changes seen in human patients are reproduced in mice injected with purified endotoxin (lipopolysaccharide, LPS). In the present study, down-regulation of genes involved in hepatic fatty acid oxidation (FAOx) and gluconeogenesis in mice exposed to LPS was challenged by nutritional intervention with Argan oil. Mice given a standard chow supplemented or not with either 6% (w/w) Argan oil (AO) or 6% (w/w) olive oil (OO) prior to exposure to LPS were explored for liver gene expressions assessed by mRNA transcript levels and/or enzyme activities. AO (or OO) food supplementation reveals that, in LPS-treated mice, hepatic expression of genes involved in FAOx and gluconeogenesis was preserved. This preventive protection might be related to the recovery of the gene expressions of nuclear receptors peroxisome proliferator-activated receptor α (PPARα) and estrogen related receptor α (ERRα) and their coactivator peroxisome proliferator-activated receptor gamma coactivator-1α, (PGC-1α). These preventive mechanisms conveyed by AO against LPS-induced metabolic dysregulation might add new therapeutic potentialities in the management of human sepsis.

11.
Mol Nutr Food Res ; 58(9): 1785-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24975132

ABSTRACT

SCOPE: Resveratrol may function as a chemopreventive agent. A recent clinical study demonstrates a reduction in tumor cell proliferation in colorectal patients receiving repeated oral ingestion of resveratrol. However, gaps remain in our knowledge of the molecular mechanisms by which resveratrol exerts its chemopreventive effect. We have previously demonstrated that resveratrol induces apoptosis in colon cancer cells and that resveratrol can sensitize chemoresistant colon cancer cells to various drugs. Based on its ability to activate peroxisome proliferator-activated receptor gamma (PPARγ) in colon cancer cells, we sought to determine the implication of this nuclear transcription factor in resveratrol-induced apoptosis. METHODS AND RESULTS: Transient transfection of cancer cells with a dominant-negative PPARγ mutant or treatment with a PPARγ antagonist (GW9662) reversed the inhibitory effect of resveratrol. Moreover, GW9662 prevented disruption of the cell cycle induced by resveratrol and consequently abrogated resveratrol-induced apoptosis. Tumor cell death was potentiated by combining resveratrol with rosiglitazone, a PPARγ agonist. CONCLUSION: The results show that PPARγ plays a role in resveratrol-induced apoptosis of colon carcinoma cells. The combination of resveratrol with a PPARγ agonist could be a promising pharmacological approach for treatment of colorectal cancer.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , PPAR gamma/metabolism , Stilbenes/pharmacology , Anilides/pharmacology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Humans , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Resveratrol , Rosiglitazone , S Phase Cell Cycle Checkpoints/drug effects , Thiazolidinediones/pharmacology
12.
Diabetes ; 63(2): 471-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24186868

ABSTRACT

Lipopolysaccharides (LPS) of the cell wall of gram-negative bacteria trigger inflammation, which is associated with marked changes in glucose metabolism. Hyperglycemia is frequently observed during bacterial infection and it is a marker of a poor clinical outcome in critically ill patients. The aim of the current study was to investigate the effect of an acute injection or continuous infusion of LPS on experimentally induced hyperglycemia in wild-type and genetically engineered mice. The acute injection of a single dose of LPS produced an increase in glucose disposal and glucose-stimulated insulin secretion (GSIS). Continuous infusion of LPS through mini-osmotic pumps was also associated with increased GSIS. Finally, manipulation of LPS detoxification by knocking out the plasma phospholipid transfer protein (PLTP) led to increased glucose disposal and GSIS. Overall, glucose tolerance and GSIS tests supported the hypothesis that mice treated with LPS develop glucose-induced hyperinsulinemia. The effects of LPS on glucose metabolism were significantly altered as a result of either the accumulation or antagonism of glucagon-like peptide 1 (GLP-1). Complementary studies in wild-type and GLP-1 receptor knockout mice further implicated the GLP-1 receptor-dependent pathway in mediating the LPS-mediated changes in glucose metabolism. Hence, enhanced GLP-1 secretion and action underlies the development of glucose-mediated hyperinsulinemia associated with endotoxemia.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Insulin/metabolism , Lipopolysaccharides/toxicity , Receptors, Glucagon/metabolism , Animals , Blood Glucose , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide-1 Receptor , Lipopolysaccharides/metabolism , Mice , Mice, Knockout , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Receptors, Glucagon/genetics
13.
Biochimie ; 98: 86-101, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24287293

ABSTRACT

The peroxisomal 3-ketoacyl-CoA thiolase B (ThB) catalyzes the thiolytic cleavage of straight chain 3-ketoacyl-CoAs. Up to now, the ability of ThB to interfere with lipid metabolism was studied in mice fed a laboratory chow enriched or not with the synthetic agonist Wy14,643, a pharmacological activator of the nuclear hormone receptor PPARα. The aim of the present study was therefore to determine whether ThB could play a role in obesity and lipid metabolism when mice are chronically fed a synthetic High Fat Diet (HFD) or a Low Fat Diet (LFD) as a control diet. To investigate this possibility, wild-type (WT) mice and mice deficient for Thb (Thb(-/-)) were subjected to either a synthetic LFD or a HFD for 25 weeks, and their responses were compared. First, when fed a normal regulatory laboratory chow, Thb(-/-) mice displayed growth retardation as well as a severe reduction in the plasma level of Growth Hormone (GH) and Insulin Growth Factor-I (IGF-I), suggesting alterations in the GH/IGF-1 pathway. When fed the synthetic diets, the corrected energy intake to body mass was significantly higher in Thb(-/-) mice, yet those mice were protected from HFD-induced adiposity. Importantly, Thb(-/-) mice also suffered from hypoglycemia, exhibited reduction in liver glycogen stores and circulating insulin levels under the LFD and the HFD. Thb deficiency was also associated with higher levels of plasma HDL (High Density Lipoproteins) cholesterol and increased liver content of cholesterol under both the LFD and the HFD. As shown by the plasma lathosterol to cholesterol ratio, a surrogate marker for cholesterol biosynthesis, whole body cholesterol de novo synthesis was increased in Thb(-/-) mice. By comparing liver RNA from WT mice and Thb(-/-) mice using oligonucleotide microarray and RT-qPCR, a coordinated decrease in the expression of critical cholesterol synthesizing genes and an increased expression of genes involved in bile acid synthesis (Cyp7a1, Cyp17a1, Akr1d1) were observed in Thb(-/-) mice. In parallel, the elevation of the lathosterol to cholesterol ratio as well as the increased expression of cholesterol synthesizing genes were observed in the kidney of Thb(-/-) mice fed the LFD and the HFD. Overall, the data indicate that ThB is not fully interchangeable with the thiolase A isoform. The present study also reveals that modulating the expression of the peroxisomal ThB enzyme can largely reverberate not only throughout fatty acid metabolism but also cholesterol, bile acid and glucose metabolism.


Subject(s)
Acetyl-CoA C-Acyltransferase/deficiency , Animals , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Cholesterol, HDL/blood , Diet, High-Fat , Dietary Fats/administration & dosage , Glucose/metabolism , Growth Hormone/blood , Insulin-Like Growth Factor I/metabolism , Intestine, Small/metabolism , Liver Glycogen/metabolism , Mice
15.
PPAR Res ; 2013: 613864, 2013.
Article in English | MEDLINE | ID: mdl-23577023

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that play pivotal roles in the regulation of a very large number of biological processes including inflammation. Using specific examples, this paper focuses on the interplay between PPARs and innate immunity/inflammation and, when possible, compares it among species. We focus on recent discoveries establishing how inflammation and PPARs interact in the context of obesity-induced inflammation and type 2 diabetes, mostly in mouse and humans. We illustrate that PPAR γ ability to alleviate obesity-associated inflammation raises an interesting pharmacologic potential. In the light of recent findings, the protective role of PPAR α and PPAR ß / δ against the hepatic inflammatory response is also addressed. While PPARs agonists are well-established agents that can treat numerous inflammatory issues in rodents and humans, surprisingly very little has been described in other species. We therefore also review the implication of PPARs in inflammatory bowel disease; acute-phase response; and central, cardiac, and endothelial inflammation and compare it along different species (mainly mouse, rat, human, and pig). In the light of the data available in the literature, there is no doubt that more studies concerning the impact of PPAR ligands in livestock should be undertaken because it may finally raise unconsidered health and sanitary benefits.

16.
Biochimie ; 93(5): 876-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21352884

ABSTRACT

Peroxisomal 3-ketoacyl-CoA thiolase B (Thb) catalyzes the final step in the peroxisomal ß-oxidation of straight-chain acyl-CoAs and is under the transcription control of the nuclear hormone receptor PPARα. PPARα binds to and is activated by the synthetic compound Wy14,643 (Wy). Here, we show that the magnitude of Wy-mediated induction of peroxisomal ß-oxidation of radiolabeled (1-(14)C) palmitate was significantly reduced in mice deficient for Thb. In contrast, mitochondrial ß-oxidation was unaltered in Thb(-/-) mice. Given that Wy-treatment induced Acox1 and MFP-1/-2 activity at a similar level in both genotypes, we concluded that the thiolase step alone was responsible for the reduced peroxisomal ß-oxidation of fatty acids. Electron microscopic analysis and cytochemical localization of catalase indicated that peroxisome proliferation in the liver after Wy-treatment was normal in Thb(-/-) mice. Intriguingly, micro-array analysis revealed that mRNA levels of genes encoding cholesterol biosynthesis enzymes were upregulated by Wy in Wild-Type (WT) mice but not in Thb(-/-) mice, which was confirmed at the protein level for the selected genes. The non-induction of genes encoding cholesterol biosynthesis enzymes by Wy in Thb(-/-) mice appeared to be unrelated to defective SREBP-2 or PPARα signaling. No difference was observed in the plasma lathosterol/cholesterol ratio (a marker for de novo cholesterol biosynthesis) between Wy-treated WT and Thb(-/-) mice, suggesting functional compensation. Overall, we conclude that ThA and SCPx/SCP2 thiolases cannot fully compensate for the absence of ThB. In addition, our data indicate that ThB is involved in the regulation of genes encoding cholesterol biosynthesis enzymes in the liver, suggesting that the peroxisome could be a promising candidate for the correction of cholesterol imbalance in dyslipidemia.


Subject(s)
Acetyl-CoA C-Acyltransferase/metabolism , Liver/enzymology , PPAR alpha/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Up-Regulation , Acetyl-CoA C-Acyltransferase/genetics , Animals , Cholesterol/genetics , Cholesterol/metabolism , Gene Deletion , Gene Expression Regulation , Hepatomegaly/genetics , Hepatomegaly/pathology , Humans , Lipid Metabolism/genetics , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Palmitates/metabolism , Peroxisome Proliferators/pharmacology , Peroxisomes/metabolism , Pyrimidines/pharmacology , Signal Transduction
17.
Biochimie ; 91(11-12): 1376-86, 2009.
Article in English | MEDLINE | ID: mdl-19772884

ABSTRACT

The peroxisomal 3-ketoacyl-CoA thiolase B (Thb) gene was previously identified as a direct target gene of PPARalpha, a nuclear hormone receptor activated by hypolipidemic fibrate drugs. To better understand the role of ThB in hepatic lipid metabolism in mice, Sv129 wild-type and Thb null mice were fed or not the selective PPARalpha agonist Wy14,643 (Wy). Here, it is shown that in contrast to some other mouse models deficient for peroxisomal enzymes, the hepatic PPARalpha signaling cascade in Thb null mice was normal under regular conditions. It is of interest that the hypotriglyceridemic action of Wy was reduced in Thb null mice underlining the conclusion that neither thiolase A nor SCPx/SCP2 thiolase can fully substitute for ThB in vivo. Moreover, a significant increased in the expression of lipogenic genes such as Stearoyl CoA Desaturase-1 (SCD1) was observed in Thb null mice fed Wy. Elevation of Scd1 mRNA and protein levels led to higher SCD1 activity, through a molecular mechanism that is probably SREBP1 independent. In agreement with higher SCD1, enrichment of liver mono-unsaturated fatty acids of the n-7 and n-9 series was found in Thb null mice fed Wy. Overall, we show that the reduced peroxisomal beta-oxidation of fat observed in Thb null mice fed Wy is associated with enhanced hepatic lipogenesis, through the combined elevation of microsomal SCD1 protein and activity. Ultimately, not only the amount but also the quality of the hepatic fatty acid pool is modulated upon the deletion of Thb.


Subject(s)
Lipid Metabolism/drug effects , PPAR alpha/antagonists & inhibitors , Peroxisomes/drug effects , Pyrimidines/pharmacology , Stearoyl-CoA Desaturase/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Fatty Acids/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Metabolism/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Microsomes, Liver/pathology , Peroxisomes/metabolism , RNA, Messenger/drug effects
18.
Endocrinology ; 148(6): 2753-63, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17347305

ABSTRACT

Recently it has become evident that obesity is associated with low-grade chronic inflammation. The transcription factor peroxisome proliferator-activated receptor alpha (PPARalpha) has been shown to have a strong antiinflammatory action in liver. However, the role of PPARalpha in obesity-induced inflammation is much less clear. Therefore, the aim of our study was to determine whether PPARalpha plays a role in obesity-induced hepatic inflammation. To induce obesity, wild-type sv129 and PPARalpha(-/-) mice were exposed to a chronic high-fat diet (HFD), using a low-fat diet (LFD) as control. In wild-type mice, HFD significantly increased the hepatic and adipose expression of numerous genes involved in inflammation. Importantly, this effect was amplified in PPARalpha(-/-) mice, suggesting an antiinflammatory role of PPARalpha in liver and adipose tissue. Further analysis identified specific chemokines and macrophage markers, including monocyte chemotactic protein 1 and F4/80(+), that were elevated in liver and adipose tissue of PPARalpha(-/-) mice, indicating increased inflammatory cell recruitment in the knockout animals. When all groups of mice were analyzed together, a significant correlation between hepatic triglycerides and expression of inflammatory markers was observed. Many inflammatory genes that were up-regulated in PPARalpha(-/-) livers by HFD were down-regulated by treatment with the PPARalpha ligand Wy-14643 under normal nonsteatotic conditions, either in vivo or in vitro, suggesting an antiinflammatory effect of PPARalpha that is independent of reduction in liver triglycerides. In conclusion, our results suggest that PPARalpha protects against obesity-induced chronic inflammation in liver by reducing hepatic steatosis, by direct down-regulation of inflammatory genes, and by attenuating inflammation in adipose tissue.


Subject(s)
Hepatitis, Animal/etiology , Hepatitis, Animal/prevention & control , Obesity/complications , PPAR alpha/physiology , Adipose Tissue/chemistry , Adipose Tissue/metabolism , Animals , Cells, Cultured , Diet, Atherogenic , Diet, Fat-Restricted , Gene Expression Profiling , Gene Expression Regulation , Hepatitis, Animal/genetics , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Knockout , Obesity/genetics , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/analysis
19.
J Hepatol ; 46(5): 869-77, 2007 May.
Article in English | MEDLINE | ID: mdl-17321000

ABSTRACT

BACKGROUND/AIMS: The Peroxisome Proliferator-Activated Receptor (PPAR) alpha belongs to the superfamily of Nuclear Receptors and plays an important role in numerous cellular processes, including lipid metabolism. It is known that PPARalpha also has an anti-inflammatory effect, which is mainly achieved by down-regulating pro-inflammatory genes. The objective of this study was to further characterize the role of PPARalpha in inflammatory gene regulation in liver. RESULTS: According to Affymetrix micro-array analysis, the expression of various inflammatory genes in liver was decreased by treatment of mice with the synthetic PPARalpha agonist Wy14643 in a PPARalpha-dependent manner. In contrast, expression of Interleukin-1 receptor antagonist (IL-1ra), which was acutely stimulated by LPS treatment, was induced by PPARalpha. Up-regulation of IL-1ra by LPS was lower in PPARalpha -/- mice compared to Wt mice. Transactivation and chromatin immunoprecipitation studies identified IL-1ra as a direct positive target gene of PPARalpha with a functional PPRE present in the promoter. Up-regulation of IL-1ra by PPARalpha was conserved in human HepG2 hepatoma cells and the human monocyte/macrophage THP-1 cell line. CONCLUSIONS: In addition to down-regulating expression of pro-inflammatory genes, PPARalpha suppresses the inflammatory response by direct up-regulation of genes with anti-inflammatory properties.


Subject(s)
Hepatitis/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Liver/metabolism , PPAR alpha/metabolism , Transcription Factors/metabolism , Animals , Anticholesteremic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Female , Gene Expression Regulation , Hepatitis/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Receptors, Interleukin-1 Type I/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Tumor Cells, Cultured , Up-Regulation/genetics
20.
J Biol Chem ; 281(2): 934-44, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16272564

ABSTRACT

Proteins secreted from adipose tissue are increasingly recognized to play an important role in the regulation of glucose metabolism. However, much less is known about their effect on lipid metabolism. The fasting-induced adipose factor (FIAF/angiopoietin-like protein 4/peroxisome proliferator-activated receptor gamma angiopoietin-related protein) was previously identified as a target of hypolipidemic fibrate drugs and insulin-sensitizing thiazolidinediones. Using transgenic mice that mildly overexpress FIAF in peripheral tissues we show that FIAF is an extremely powerful regulator of lipid metabolism and adiposity. FIAF overexpression caused a 50% reduction in adipose tissue weight, partly by stimulating fatty acid oxidation and uncoupling in fat. In addition, FIAF overexpression increased plasma levels of triglycerides, free fatty acids, glycerol, total cholesterol, and high density lipoprotein (HDL)-cholesterol. Functional tests indicated that FIAF overexpression severely impaired plasma triglyceride clearance but had no effect on very low density lipoprotein production. The effects of FIAF overexpression were amplified by a high fat diet, resulting in markedly elevated plasma and liver triglycerides, plasma free fatty acids, and plasma glycerol levels, and impaired glucose tolerance in FIAF transgenic mice fed a high fat diet. Remarkably, in mice the full-length form of FIAF was physically associated with HDL, whereas truncated FIAF was associated with low density lipoprotein. In human both full-length and truncated FIAF were associated with HDL. The composite data suggest that via physical association with plasma lipoproteins, FIAF acts as a powerful signal from fat and other tissues to prevent fat storage and stimulate fat mobilization. Our data indicate that disturbances in FIAF signaling might be involved in dyslipidemia.


Subject(s)
Adipose Tissue/metabolism , Blood Proteins/genetics , Blood Proteins/physiology , Lipoproteins/chemistry , Angiopoietin-Like Protein 4 , Angiopoietins , Animals , Blood Proteins/chemistry , Body Weight , Cholesterol/metabolism , Fats/chemistry , Gene Expression , Glucose/metabolism , Glucose Tolerance Test , Humans , Hypercholesterolemia/metabolism , Immunoblotting , Insulin/metabolism , Lipase/metabolism , Lipids/chemistry , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/chemistry , Lipoproteins, VLDL/chemistry , Liver/enzymology , Male , Mice , Mice, Transgenic , Models, Genetic , Oligonucleotide Array Sequence Analysis , Protein Binding , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Triglycerides/blood , Triglycerides/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...