Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Int J Obes (Lond) ; 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117851

RÉSUMÉ

BACKGROUND/OBJECTIVES: Melanocortins mediate their biological functions via five different melanocortin receptors (MC1R - MC5R). MC1R is expressed in the skin and leukocytes, where it regulates skin pigmentation and inflammatory responses. MC1R is also present in the liver and white adipose tissue, but its functional role in these tissues is unclear. This study aimed at determining the regulatory role of MC1R in fatty acid metabolism. METHODS: Male recessive yellow (Mc1re/e) mice, a model of global MC1R deficiency, and male hepatocyte-specific MC1R deficient mice (Mc1r LKO) were fed a chow or Western diet for 12 weeks. The mouse models were characterized for body weight and composition, liver adiposity, adipose tissue mass and morphology, glucose metabolism and lipid metabolism. Furthermore, qPCR and RNA sequencing analyses were used to investigate gene expression profiles in the liver and adipose tissue. HepG2 cells and primary mouse hepatocytes were used to study the effects of pharmacological MC1R activation. RESULTS: Chow- and Western diet-fed Mc1re/e showed increased liver weight, white adipose tissue mass and plasma triglyceride (TG) concentration compared to wild type mice. This phenotype occurred without significant changes in food intake, body weight, physical activity or glucose metabolism. Mc1r LKO mice displayed a similar phenotype characterized by larger fat depots, increased adipocyte hypertrophy and enhanced accumulation of TG in the liver and plasma. In terms of gene expression, markers of de novo lipogenesis, inflammation and apoptosis were upregulated in the liver of Mc1r LKO mice, while enzymes regulating lipolysis were downregulated in white adipose tissue of these mice. In cultured hepatocytes, selective activation of MC1R reduced ChREBP expression, which is a central transcription factor for lipogenesis. CONCLUSIONS: Hepatocyte-specific loss of MC1R disturbs fatty acid metabolism in the liver and leads to an obesity phenotype characterized by enhanced adipocyte hypertrophy and TG accumulation in the liver and circulation.

2.
Front Immunol ; 15: 1394420, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979417

RÉSUMÉ

Background: Mobilization of certain immune cells may improve the ability of the immune system to combat tumor cells, but the effect of acute exercise on mobilizing immune cells has been sparsely investigated in cancer patients. Therefore, we examined how acute exercise influences circulating immune cells in breast cancer patients. Methods: Nineteen newly diagnosed breast cancer patients aged 36-68 performed 30 minutes of moderate-intensity exercise with a cycle ergometer. Blood samples were collected at various time points: at rest, at 15 (E15) and 30 minutes (E30) after onset of the exercise, and at 30 and 60 minutes post-exercise. We analyzed several immune cell subsets using flow cytometry. Results: Acute exercise increased the number of total leukocytes, neutrophils, lymphocytes, monocytes, basophils, total T-cells, CD4+ T-cells, T helper (Th) 2-cells, Th 17-cells, CD8+ T-cells, CD4-CD8- T-cells, CD56+ natural killer (NK) cells, and CD14-CD16+ monocytes. Many of the changes were transient. Proportions of NK-cells and CD8+ T-cells increased, while the proportion of myeloid derived suppressor cells (MDSCs) reduced, and proportion of regulatory T-cells remained unchanged by exercise. Several associations were detected between cell mobilizations and disease state. For instance, tumor size correlated negatively with NK cell mobilization at E15, and progesterone receptor positivity correlated negatively with CD8+ T-cell mobilization. Conclusion: The findings show that the proportions of CD8+ T-cells and NK cells increased and the proportion of MDSCs proportion decreased in breast cancer patients after 30-minute exercise, suggesting a change in the profile of circulating immune cells towards more cytotoxic/anti-tumorigenic. The mobilization of some immune cells also appears to be related to the disease state.


Sujet(s)
Tumeurs du sein , Exercice physique , Cellules tueuses naturelles , Humains , Femelle , Tumeurs du sein/immunologie , Adulte d'âge moyen , Adulte , Sujet âgé , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme
3.
EMBO Rep ; 25(4): 1987-2014, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38454158

RÉSUMÉ

α-Melanocyte-stimulating hormone (α-MSH) regulates diverse physiological functions by activating melanocortin receptors (MC-R). However, the role of α-MSH and its possible target receptors in the heart remain completely unknown. Here we investigate whether α-MSH could be involved in pathological cardiac remodeling. We found that α-MSH was highly expressed in the mouse heart with reduced ventricular levels after transverse aortic constriction (TAC). Administration of a stable α-MSH analog protected mice against TAC-induced cardiac hypertrophy and systolic dysfunction. In vitro experiments revealed that MC5-R in cardiomyocytes mediates the anti-hypertrophic signaling of α-MSH. Silencing of MC5-R in cardiomyocytes induced hypertrophy and fibrosis markers in vitro and aggravated TAC-induced cardiac hypertrophy and fibrosis in vivo. Conversely, pharmacological activation of MC5-R improved systolic function and reduced cardiac fibrosis in TAC-operated mice. In conclusion, α-MSH is expressed in the heart and protects against pathological cardiac remodeling by activating MC5-R in cardiomyocytes. These results suggest that analogs of naturally occurring α-MSH, that have been recently approved for clinical use and have agonistic activity at MC5-R, may be of benefit in treating heart failure.


Sujet(s)
Remodelage ventriculaire , Hormone mélanotrope alpha , Souris , Animaux , Hormone mélanotrope alpha/pharmacologie , Récepteur corticotrophine , Récepteurs à la mélanocortine , Cardiomégalie/génétique , Fibrose
4.
Elife ; 122023 07 25.
Article de Anglais | MEDLINE | ID: mdl-37490042

RÉSUMÉ

Melanocortin 1 receptor (MC1-R) is widely expressed in melanocytes and leukocytes and is thus strongly implicated in the regulation of skin pigmentation and inflammation. MC1-R has also been found in the rat and human liver, but its functional role has remained elusive. We hypothesized that MC1-R is functionally active in the liver and involved in the regulation of cholesterol and bile acid metabolism. We generated hepatocyte-specific MC1-R knock-out (Mc1r LKO) mice and phenotyped the mouse model for lipid profiles, liver histology, and bile acid levels. Mc1r LKO mice had significantly increased liver weight, which was accompanied by elevated levels of total cholesterol and triglycerides in the liver as well as in the plasma. These mice demonstrated also enhanced liver fibrosis and a disturbance in bile acid metabolism as evidenced by markedly reduced bile acid levels in the plasma and feces. Mechanistically, using HepG2 cells as an in vitro model, we found that selective activation of MC1-R in HepG2 cells reduced cellular cholesterol content and enhanced uptake of low- and high-density lipoprotein particles via a cAMP-independent mechanism. In conclusion, the present results demonstrate that MC1-R signaling in hepatocytes regulates cholesterol and bile acid metabolism and its deficiency leads to hypercholesterolemia and enhanced lipid accumulation and fibrosis in the liver.


Sujet(s)
Foie , Récepteur de la mélanocortine de type 1 , Humains , Souris , Rats , Animaux , Cholestérol , Hépatocytes , Acides et sels biliaires
6.
Am J Pathol ; 193(8): 1072-1080, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37236505

RÉSUMÉ

The incidence of nonalcoholic fatty liver disease is a continuously growing health problem worldwide, along with obesity. Therefore, novel methods to both efficiently study the manifestation of nonalcoholic fatty liver disease and to analyze drug efficacy in preclinical models are needed. The present study developed a deep neural network-based model to quantify microvesicular and macrovesicular steatosis in the liver on hematoxylin-eosin-stained whole slide images, using the cloud-based platform, Aiforia Create. The training data included a total of 101 whole slide images from dietary interventions of wild-type mice and from two genetically modified mouse models with steatosis. The algorithm was trained for the following: to detect liver parenchyma, to exclude the blood vessels and any artefacts generated during tissue processing and image acquisition, to recognize and differentiate the areas of microvesicular and macrovesicular steatosis, and to quantify the recognized tissue area. The results of the image analysis replicated well the evaluation by expert pathologists and correlated well with the liver fat content measured by EchoMRI ex vivo, and the correlation with total liver triglycerides was notable. In conclusion, the developed deep learning-based model is a novel tool for studying liver steatosis in mouse models on paraffin sections and, thus, can facilitate reliable quantification of the amount of steatosis in large preclinical study cohorts.


Sujet(s)
Apprentissage profond , Stéatose hépatique non alcoolique , Souris , Animaux , Stéatose hépatique non alcoolique/imagerie diagnostique , Foie , 29935 , Algorithmes , Modèles animaux de maladie humaine
7.
Sci Rep ; 13(1): 6561, 2023 04 21.
Article de Anglais | MEDLINE | ID: mdl-37085562

RÉSUMÉ

The role of exercise in cancer prevention and control is increasingly recognized, and based on preclinical studies, it is hypothesized that mobilization of leukocytes plays an important role in the anti-tumor effect. Thus, we examined how 10-min acute exercise modulates immune cells in newly diagnosed breast cancer patients. Blood samples were taken at rest, immediately after exercise and 30 min after exercise and phenotypic characterization of major leukocyte subsets was done using 9-color flow cytometry. Total leukocyte count increased by 29%, CD8+ T cell count by 34%, CD19+ B cell count by 18%, CD56+CD16+ NK cell count by 130%, and CD14+CD16+ monocyte count by 51% immediately after acute exercise. Mobilization of CD45+, CD8+, CD19+, and CD56+CD16+ cells correlated positively with exercising systolic blood pressure, heart rate percentage of age predicted maximal heart rate, rate pressure product, and mean arterial pressure. Our findings indicate that a single bout of acute exercise of only 10 min can cause leukocytosis in breast cancer patients. Mobilization of leukocytes appear to be directly related to the intensity of exercise. It is possible that the positive effect of exercise on oncologic outcome might be partly due to immune cell mobilization as documented in the present study.


Sujet(s)
Tumeurs du sein , Humains , Femelle , Leucocytes , Numération des leucocytes , Cellules tueuses naturelles , Exercice physique/physiologie
8.
Nat Cardiovasc Res ; 1: 1056-1071, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36523570

RÉSUMÉ

Dissecting the pathways regulating the adaptive immune response in atherosclerosis is of particular therapeutic interest. Here we report that the lipid G-protein coupled receptor GPR55 is highly expressed by splenic plasma cells (PC), upregulated in mouse spleens during atherogenesis and human unstable or ruptured compared to stable plaques. Gpr55-deficient mice developed larger atherosclerotic plaques with increased necrotic core size compared to their corresponding controls. Lack of GPR55 hyperactivated B cells, disturbed PC maturation and resulted in immunoglobulin (Ig)G overproduction. B cell-specific Gpr55 depletion or adoptive transfer of Gpr55-deficient B cells was sufficient to promote plaque development and elevated IgG titers. In vitro, the endogenous GPR55 ligand lysophsophatidylinositol (LPI) enhanced PC proliferation, whereas GPR55 antagonism blocked PC maturation and increased their mitochondrial content. Collectively, these discoveries provide previously undefined evidence for GPR55 in B cells as a key modulator of the adaptive immune response in atherosclerosis.

9.
Front Physiol ; 13: 1078512, 2022.
Article de Anglais | MEDLINE | ID: mdl-36714311

RÉSUMÉ

Background: Studies have shown that acute exercise can mobilize several leukocyte subpopulations in healthy individuals. Our aim was to investigate whether a 10-min acute exercise has an effect on immune cell proportions in lymphoma patients. Methods: This study included seven lymphoma patients referred to curative oncologic therapy. Three had Hodgkin and four non-Hodgkin lymphoma, one was female, and their mean age was 51. Patients underwent a 10-min acute exercise on a bicycle ergometer at moderate exercise intensity. Whole blood samples were taken at rest, immediately after exercise, and 30 min after exercise. Leukocyte subpopulation levels were determined using flow cytometry. Results: Proportions of total NK cells and CD56+CD16+ NK cells of total leukocytes increased immediately after exercise and decreased back to baseline at 30 min post-exercise. Proportion of CD8+ T cells of total T cells increased and proportion of CD4+ T cells of total T cells decreased immediately after exercise, and both returned to baseline at 30 min post-exercise. There was no change in the proportions of B cells, granulocytes, or monocytes. Exercising diastolic blood pressure correlated positively with changes in total NK cell and CD56+CD16+ NK cell proportions, and exercising mean arterial pressure correlated positively with change in CD56+CD16+ NK cell proportion. Conclusion: Our findings indicate that a single acute exercise bout of only 10 min can cause leukocytosis in lymphoma patients, particularly on cytotoxic T cells and NK cells, which are the most important immune cells fighting against cancer.

10.
Front Immunol ; 12: 774013, 2021.
Article de Anglais | MEDLINE | ID: mdl-34868038

RÉSUMÉ

Melanocortin receptor 1 (MC1-R) is expressed in leukocytes, where it mediates anti-inflammatory actions. We have previously observed that global deficiency of MC1-R signaling perturbs cholesterol homeostasis, increases arterial leukocyte accumulation and accelerates atherosclerosis in apolipoprotein E knockout (Apoe-/-) mice. Since various cell types besides leukocytes express MC1-R, we aimed at investigating the specific contribution of leukocyte MC1-R to the development of atherosclerosis. For this purpose, male Apoe-/- mice were irradiated, received bone marrow from either female Apoe-/- mice or MC1-R deficient Apoe-/- mice (Apoe-/- Mc1re/e) and were analyzed for tissue leukocyte profiles and atherosclerotic plaque phenotype. Hematopoietic MC1-R deficiency significantly elevated total leukocyte counts in the blood, bone marrow and spleen, an effect that was amplified by feeding mice a cholesterol-rich diet. The increased leukocyte counts were largely attributable to expanded lymphocyte populations, particularly CD4+ T cells. Furthermore, the number of monocytes was elevated in Apoe-/- Mc1re/e chimeric mice and it paralleled an increase in hematopoietic stem cell count in the bone marrow. Despite robust leukocytosis, atherosclerotic plaque size and composition as well as arterial leukocyte counts were unaffected by MC1-R deficiency. To address this discrepancy, we performed an in vivo homing assay and found that MC1-R deficient CD4+ T cells and monocytes were preferentially entering the spleen rather than homing in peri-aortic lymph nodes. This was mechanistically associated with compromised chemokine receptor 5 (CCR5)-dependent migration of CD4+ T cells and a defect in the recycling capacity of CCR5. Finally, our data demonstrate for the first time that CD4+ T cells also express MC1-R. In conclusion, MC1-R regulates hematopoietic stem cell proliferation and tissue leukocyte counts but its deficiency in leukocytes impairs cell migration via a CCR5-dependent mechanism.


Sujet(s)
Athérosclérose/étiologie , Athérosclérose/métabolisme , Cellules sanguines/métabolisme , Prédisposition aux maladies , Leucocytes/métabolisme , Récepteur de la mélanocortine de type 1/déficit , Animaux , Athérosclérose/anatomopathologie , Marqueurs biologiques , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Cytokines/métabolisme , Modèles animaux de maladie humaine , Immunophénotypage , Médiateurs de l'inflammation/métabolisme , Leucocytes/anatomopathologie , Souris , Souris knockout
11.
Eur J Pharmacol ; 880: 173186, 2020 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-32416182

RÉSUMÉ

The melanocortin MC1 and MC3 receptors elicit anti-inflammatory actions in leukocytes and activation of these receptors has been shown to alleviate arterial inflammation in experimental atherosclerosis. Thus, we aimed to investigate whether selective targeting of melanocortin MC3 receptor protects against atherosclerosis. Apolipoprotein E deficient (ApoE-/-) mice were fed high-fat diet for 12 weeks and randomly assigned to receive either vehicle (n = 11) or the selective melanocortin MC3 receptor agonist [D-Trp(8)]-gamma-melanocyte-stimulating hormone ([D-Trp8]-γ-MSH; 15 µg/day, n = 10) for the last 4 weeks. Lesion size as well as macrophage and collagen content in the aortic root plaques were determined. Furthermore, leukocyte counts in the blood and aorta and cytokine mRNA expression levels in the spleen, liver and aorta were quantified. No effect was observed in the body weight development or plasma cholesterol level between the two treatment groups. However, [D-Trp8]-γ-MSH treatment significantly reduced plasma levels of chemokine (C-C motif) ligands 2, 4 and 5. Likewise, cytokine and adhesion molecule expression levels were reduced in the spleen and liver of γ-MSH-treated mice, but not substantially in the aorta. In line with these findings, [D-Trp8]-γ-MSH treatment reduced leukocyte counts in the blood and aorta. Despite reduced inflammation, [D-Trp8]-γ-MSH did not change lesion size, macrophage content or collagen deposition of aortic root plaques. In conclusion, the findings indicate that selective activation of melanocortin MC3 receptor by [D-Trp8]-γ-MSH suppresses systemic and local inflammation and thereby also limits leukocyte accumulation in the aorta. However, the treatment was ineffective in reducing atherosclerotic plaque size.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Hormones mélanotropes/usage thérapeutique , Plaque d'athérosclérose/traitement médicamenteux , Récepteur de la mélanocortine de type 3/agonistes , Animaux , Anti-inflammatoires/pharmacologie , Aorte/effets des médicaments et des substances chimiques , Aorte/immunologie , Aorte/anatomopathologie , Cellules cultivées , Cholestérol/sang , Cytokines/sang , Cytokines/génétique , Alimentation riche en graisse , Cellules endothéliales , Femelle , Inflammation/immunologie , Numération des leucocytes , Foie/effets des médicaments et des substances chimiques , Foie/immunologie , Hormones mélanotropes/pharmacologie , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose/immunologie , Plaque d'athérosclérose/anatomopathologie , Récepteur de la mélanocortine de type 3/immunologie , Rate/effets des médicaments et des substances chimiques , Rate/immunologie
12.
JACC Basic Transl Sci ; 5(4): 360-373, 2020 Apr.
Article de Anglais | MEDLINE | ID: mdl-32368695

RÉSUMÉ

This study showed that treatment with a therapeutic monoclonal immunoglobulin-G1 antibody against phosphorylcholine on oxidized phospholipids preserves coronary flow reserve and attenuates atherosclerotic inflammation as determined by the uptake of 18F-fluorodeoxyglucose in atherosclerotic mice. The noninvasive imaging techniques represent translational tools to assess the efficacy of phosphorylcholine-targeted therapy on coronary artery function and atherosclerosis in clinical studies.

14.
Sci Rep ; 8(1): 15078, 2018 10 10.
Article de Anglais | MEDLINE | ID: mdl-30305673

RÉSUMÉ

α-melanocyte-stimulating hormone (α-MSH) is processed from pro-opiomelanocortin (POMC) and mediates anti-inflammatory actions in leukocytes. α-MSH also promotes macrophage reverse cholesterol transport by inducing ATP-binding cassette transporters ABCA1 and ABCG1. Here we investigated the regulation of POMC and α-MSH expression in atherosclerosis. First, transcript levels of POMC and its processing enzymes were analyzed in human arterial plaques (n = 68) and non-atherosclerotic controls (n = 24) as well as in whole blood samples from coronary artery disease patients (n = 55) and controls (n = 45) by microarray. POMC expression was increased in femoral plaques compared to control samples as well as in unstable advanced plaques. α-MSH-producing enzyme, carboxypeptidase E, was down-regulated, whereas prolylcarboxypeptidase, an enzyme inactivating α-MSH, was up-regulated in unstable plaques compared to stable plaques, suggesting a possible reduction in intraplaque α-MSH levels. Second, immunohistochemical analyses revealed the presence of α-MSH in atherosclerotic plaques and its localization in macrophages and other cell types. Lastly, supporting the role of α-MSH in reverse cholesterol transport, POMC expression correlated with ABCA1 and ABCG1 in human plaque and whole blood samples. In conclusion, α-MSH is expressed in atherosclerotic plaques and its processing enzymes associate with plaque stability, suggesting that measures to enhance the local bioavailability of α-MSH might protect against atherosclerosis.


Sujet(s)
Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Pro-opiomélanocortine/métabolisme , Animaux , Athérosclérose/génétique , Transport biologique , Études cas-témoins , Cholestérol/métabolisme , Modèles animaux de maladie humaine , Technique d'immunofluorescence , Expression des gènes , Analyse de profil d'expression de gènes , Humains , Immunohistochimie , Souris , Souris knockout , Plaque d'athérosclérose/génétique , Plaque d'athérosclérose/anatomopathologie , Protéolyse , Hormone mélanotrope alpha/métabolisme
15.
Arterioscler Thromb Vasc Biol ; 38(11): 2562-2575, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30354245

RÉSUMÉ

Objective- Palmitoylethanolamide is an endogenous fatty acid mediator that is synthetized from membrane phospholipids by N-acyl phosphatidylethanolamine phospholipase D. Its biological actions are primarily mediated by PPAR-α (peroxisome proliferator-activated receptors α) and the orphan receptor GPR55. Palmitoylethanolamide exerts potent anti-inflammatory actions but its physiological role and promise as a therapeutic agent in chronic arterial inflammation, such as atherosclerosis remain unexplored. Approach and Results- First, the polarization of mouse primary macrophages towards a proinflammatory phenotype was found to reduce N-acyl phosphatidylethanolamine phospholipase D expression and palmitoylethanolamide bioavailability. N-acyl phosphatidylethanolamine phospholipase D expression was progressively downregulated in the aorta of apolipoprotein E deficient (ApoE-/-) mice during atherogenesis. N-acyl phosphatidylethanolamine phospholipase D mRNA levels were also downregulated in unstable human plaques and they positively associated with smooth muscle cell markers and negatively with macrophage markers. Second, ApoE-/- mice were fed a high-fat diet for 4 or 16 weeks and treated with either vehicle or palmitoylethanolamide (3 mg/kg per day, 4 weeks) to study the effects of palmitoylethanolamide on early established and pre-established atherosclerosis. Palmitoylethanolamide treatment reduced plaque size in early atherosclerosis, whereas in pre-established atherosclerosis, palmitoylethanolamide promoted signs of plaque stability as evidenced by reduced macrophage accumulation and necrotic core size, increased collagen deposition and downregulation of M1-type macrophage markers. Mechanistically, we found that palmitoylethanolamide, by activating GPR55, increases the expression of the phagocytosis receptor MerTK (proto-oncogene tyrosine-protein kinase MER) and enhances macrophage efferocytosis, indicative of proresolving properties. Conclusions- The present study demonstrates that palmitoylethanolamide protects against atherosclerosis by promoting an anti-inflammatory and proresolving phenotype of lesional macrophages, representing a new therapeutic approach to resolve arterial inflammation.


Sujet(s)
Anti-inflammatoires/pharmacologie , Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Éthanolamines/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Acides palmitiques/pharmacologie , Phagocytose/effets des médicaments et des substances chimiques , Plaque d'athérosclérose , Amides , Animaux , Aorte/métabolisme , Aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/métabolisme , Maladies de l'aorte/anatomopathologie , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Lignée cellulaire , Modèles animaux de maladie humaine , Femelle , Humains , Macrophages/métabolisme , Macrophages/anatomopathologie , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Phénotype , Phospholipase D/métabolisme , Proto-oncogène Mas , Rats , Récepteurs de cannabinoïdes/génétique , Récepteurs de cannabinoïdes/métabolisme , Facteurs temps , c-Mer Tyrosine kinase/métabolisme
16.
Arterioscler Thromb Vasc Biol ; 38(5): 1007-1019, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29567680

RÉSUMÉ

OBJECTIVE: Cardiovascular diseases and depression are the leading causes of disability in Western countries. Clinical data on potential cardiovascular effects of serotonin reuptake inhibitors (SSRIs), the most commonly used antidepressant drugs, are controversial. In addition to blocking serotonin reuptake transporter in the brain, SSRIs deplete the major peripheral serotonin (5-hydroxytryptamine [5-HT]) storage by inhibiting serotonin reuptake transporter-mediated uptake in platelets. In this study, we aimed to investigate the effect of chronic SSRI intake on the development of atherosclerosis. APPROACH AND RESULTS: Treatment of apolipoprotein E-deficient mice with the SSRI fluoxetine for 2, 4, or 16 weeks increased atherosclerotic lesion formation, with most pronounced effect during early plaque development. Intravital microscopy of carotid arteries revealed enhanced myeloid cell adhesion on fluoxetine treatment. Mechanistically, we found that fluoxetine augmented vascular permeability and increased chemokine-induced integrin-binding activity of circulating leukocytes. In vitro stimulation of murine blood demonstrated that fluoxetine, but not 5-HT, could directly promote ß1 and ß2 integrin activation provided C-C motif chemokine ligand 5 was also present. Similar effects were observed with the SSRI escitalopram. Enhanced C-C motif chemokine ligand 5-induced integrin activation by fluoxetine was also confirmed in a human neutrophil-like cell line. In contrast to the proatherogenic properties of fluoxetine, pharmacological inhibition of the peripheral 5-HT synthesizing enzyme tryptophan hydroxylase 1 did not promote atherosclerosis, suggesting that the proatherogenic effect of fluoxetine occurs independent of peripheral 5-HT depletion. CONCLUSIONS: SSRI intake may promote atherosclerosis and therefore potentially increase the risk for acute cardiovascular events by a mechanism that is independent of 5-HT depletion.


Sujet(s)
Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/induit chimiquement , Athérosclérose/induit chimiquement , Artères carotides/effets des médicaments et des substances chimiques , Artériopathies carotidiennes/induit chimiquement , Fluoxétine/toxicité , Plaque d'athérosclérose , Inbiteurs sélectifs de la recapture de la sérotonine/toxicité , Animaux , Aorte/métabolisme , Aorte/anatomopathologie , Maladies de l'aorte/sang , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/sang , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Antigènes CD18/sang , Perméabilité capillaire/effets des médicaments et des substances chimiques , Artères carotides/métabolisme , Artères carotides/anatomopathologie , Artériopathies carotidiennes/sang , Artériopathies carotidiennes/génétique , Artériopathies carotidiennes/anatomopathologie , Adhérence cellulaire/effets des médicaments et des substances chimiques , Chimiokine CCL5/sang , Modèles animaux de maladie humaine , Évolution de la maladie , Calendrier d'administration des médicaments , Fluoxétine/administration et posologie , Cellules HEK293 , Cellules HL-60 , Humains , Antigènes CD29/sang , Mâle , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Cellules myéloïdes/effets des médicaments et des substances chimiques , Cellules myéloïdes/métabolisme , Sérotonine/sang , Inbiteurs sélectifs de la recapture de la sérotonine/administration et posologie , Transduction du signal , Facteurs temps
17.
J Endocrinol ; 236(3): 111-123, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29317531

RÉSUMÉ

Atherosclerosis is a chronic inflammatory disease of the arteries. The disease is initiated by endothelial dysfunction that allows the transport of leukocytes and low-density lipoprotein into the vessel wall forming atherosclerotic plaques. The melanocortin system is an endogenous peptide system that regulates, for example, energy homeostasis and cardiovascular function. Melanocortin treatment with endogenous or synthetic melanocortin peptides reduces body weight, protects the endothelium and alleviates vascular inflammation, but the long-term effects of melanocortin system activation on atheroprogression remain largely unknown. In this study, we evaluated the effects of transgenic melanocortin overexpression in a mouse model of atherosclerosis. Low-density lipoprotein receptor-deficient mice overexpressing alpha- and gamma3-MSH (MSH-OE) and their wild-type littermates were fed either a regular chow or Western-style diet for 16 weeks. During this time, their metabolic parameters were monitored. The aortae were collected for functional analysis, and the plaques in the aortic root and arch were characterised by histological and immunohistochemical stainings. The aortic expression of inflammatory mediators was determined by quantitative PCR. We found that transgenic MSH-OE improved glucose tolerance and limited atherosclerotic plaque formation particularly in Western diet-fed mice. In terms of aortic vasoreactivity, MSH-OE blunted alpha1-adrenoceptor-mediated vasoconstriction and enhanced relaxation response to acetylcholine, indicating improved endothelial function. In addition, MSH-OE markedly attenuated Western diet-induced upregulation of proinflammatory cytokines (Ccl2, Ccl5 and Il6) that contribute to the pathogenesis of atherosclerosis. These results show that the activation of the melanocortin system improves glucose homeostasis and limits diet-induced vascular inflammation and atherosclerotic plaque formation.


Sujet(s)
Athérosclérose/prévention et contrôle , Régime occidental/effets indésirables , Inflammation/prévention et contrôle , Mélanocortines/physiologie , Récepteurs aux lipoprotéines LDL/déficit , Animaux , Aorte/métabolisme , Aorte/anatomopathologie , Cytokines/génétique , Femelle , Expression des gènes , Intolérance au glucose/prévention et contrôle , Homéostasie/physiologie , Immunohistochimie , Inflammation/physiopathologie , Mâle , Mélanocortines/génétique , Souris , Souris knockout , Souris transgéniques , Plaque d'athérosclérose/anatomopathologie , Récepteurs aux lipoprotéines LDL/génétique , Vasoconstriction , Vasodilatation , Hormone mélanotrope alpha/génétique , Hormone mélanotrope gamma/génétique
18.
Arterioscler Thromb Vasc Biol ; 38(2): 313-323, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-29284608

RÉSUMÉ

OBJECTIVE: The MC1-R (melanocortin 1 receptor) is expressed by monocytes and macrophages where it mediates anti-inflammatory actions. MC1-R also protects against macrophage foam cell formation primarily by promoting cholesterol efflux through the ABCA1 (ATP-binding cassette transporter subfamily A member 1) and ABCG1 (ATP-binding cassette transporter subfamily G member 1). In this study, we aimed to investigate whether global deficiency in MC1-R signaling affects the development of atherosclerosis. APPROACH AND RESULTS: Apoe-/- (apolipoprotein E deficient) mice were crossed with recessive yellow (Mc1re/e) mice carrying dysfunctional MC1-R and fed a high-fat diet to induce atherosclerosis. Apoe-/- Mc1re/e mice developed significantly larger atherosclerotic lesions in the aortic sinus and in the whole aorta compared with Apoe-/- controls. In terms of plaque composition, MC1-R deficiency was associated with less collagen and smooth muscle cells and increased necrotic core, indicative of more vulnerable lesions. These changes were accompanied by reduced Abca1 and Abcg1 expression in the aorta. Furthermore, Apoe-/- Mc1re/e mice showed a defect in bile acid metabolism that aggravated high-fat diet-induced hypercholesterolemia and hepatic lipid accumulation. Flow cytometric analysis of leukocyte profile revealed that dysfunctional MC1-R enhanced arterial accumulation of classical Ly6Chigh monocytes and macrophages, effects that were evident in mice fed a normal chow diet but not under high-fat diet conditions. In support of enhanced arterial recruitment of Ly6Chigh monocytes, these cells had increased expression of L-selectin and P-selectin glycoprotein ligand 1. CONCLUSIONS: The present study highlights the importance of MC1-R in the development of atherosclerosis. Deficiency in MC1-R signaling exacerbates atherosclerosis by disturbing cholesterol handling and by increasing arterial monocyte accumulation.


Sujet(s)
Aorte/métabolisme , Maladies de l'aorte/métabolisme , Athérosclérose/métabolisme , Souris invalidées pour les gènes ApoE , Monocytes/métabolisme , Plaque d'athérosclérose , Récepteur de la mélanocortine de type 1/déficit , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/métabolisme , Membre-1 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Animaux , Aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Cellules cultivées , Cholestérol/métabolisme , Alimentation riche en graisse , Modèles animaux de maladie humaine , Stéatose hépatique/génétique , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Hypercholestérolémie/génétique , Hypercholestérolémie/métabolisme , Hypercholestérolémie/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Mâle , Souris de lignée C57BL , Monocytes/anatomopathologie , Récepteur de la mélanocortine de type 1/génétique
19.
Circulation ; 136(1): 83-97, 2017 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-28450348

RÉSUMÉ

BACKGROUND: The melanocortin 1 receptor (MC1-R) is expressed by monocytes and macrophages, where it exerts anti-inflammatory actions on stimulation with its natural ligand α-melanocyte-stimulating hormone. The present study was designed to investigate the specific role of MC1-R in the context of atherosclerosis and possible regulatory pathways of MC1-R beyond anti-inflammation. METHODS: Human and mouse atherosclerotic samples and primary mouse macrophages were used to study the regulatory functions of MC1-R. The impact of pharmacological MC1-R activation on atherosclerosis was assessed in apolipoprotein E-deficient mice. RESULTS: Characterization of human and mouse atherosclerotic plaques revealed that MC1-R expression localizes in lesional macrophages and is significantly associated with the ATP-binding cassette transporters ABCA1 and ABCG1, which are responsible for initiating reverse cholesterol transport. Using bone marrow-derived macrophages, we observed that α-melanocyte-stimulating hormone and selective MC1-R agonists similarly promoted cholesterol efflux, which is a counterregulatory mechanism against foam cell formation. Mechanistically, MC1-R activation upregulated the levels of ABCA1 and ABCG1. These effects were accompanied by a reduction in cell surface CD36 expression and in cholesterol uptake, further protecting macrophages from excessive lipid accumulation. Conversely, macrophages deficient in functional MC1-R displayed a phenotype with impaired efflux and enhanced uptake of cholesterol. Pharmacological targeting of MC1-R in atherosclerotic apolipoprotein E-deficient mice reduced plasma cholesterol levels and aortic CD36 expression and increased plaque ABCG1 expression and signs of plaque stability. CONCLUSIONS: Our findings identify a novel role for MC1-R in macrophage cholesterol transport. Activation of MC1-R confers protection against macrophage foam cell formation through a dual mechanism: It prevents cholesterol uptake while concomitantly promoting ABCA1- and ABCG1-mediated reverse cholesterol transport.


Sujet(s)
Cholestérol/métabolisme , Macrophages/métabolisme , Récepteur de la mélanocortine de type 1/métabolisme , Transduction du signal/physiologie , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Transport biologique/physiologie , Femelle , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Cellules HEK293 , Humains , Macrophages/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Souris knockout , Répartition aléatoire , Récepteur de la mélanocortine de type 1/agonistes , Transduction du signal/effets des médicaments et des substances chimiques , Hormone mélanotrope alpha/métabolisme , Hormone mélanotrope alpha/pharmacologie
20.
Thromb Haemost ; 117(1): 164-175, 2017 01 05.
Article de Anglais | MEDLINE | ID: mdl-27786334

RÉSUMÉ

The co-stimulatory molecule CD70 is expressed on activated immune cells and is known to modulate responses of T, B, and NK cells via its receptor CD27. Until now, there is only limited data describing the role of CD70 in atherosclerosis. We observed that ruptured human carotid atherosclerotic plaques displayed higher CD70 expression than stable carotid atherosclerotic plaques, and that CD70 expression in murine atheroma localized to macrophages. Lack of CD70 impaired the inflammatory capacity (e. g. reactive oxygen species and nitric oxide production) of bone marrow-derived macrophages, increased both M1-like and M2-like macrophage markers, and rendered macrophages metabolically inactive and prone to apoptosis. Moreover, CD70-deficient macrophages expressed diminished levels of scavenger receptors and ABC-transporters, impairing uptake of oxidised low-density lipoprotein (oxLDL) and cholesterol efflux, respectively. Hyperlipidaemic Apoe-/- mice reconstituted with CD70-deficient bone marrow displayed a profound increase in necrotic core size, plaque area, and number of lesional macrophages as compared to mice receiving control bone marrow. Accordingly, 18 week-old, chow diet-fed CD70-deficient Apoe-/- mice displayed larger atheroma characterised by lower cellularity and more advanced plaque phenotype than Apoe-/- mice. In conclusion, CD70 promotes macrophage function and viability and is crucial for effective phagocytosis and efflux of oxLDL. Deficiency in CD70 results in more advanced atheroma. Our data suggest that CD70 mitigates atherosclerosis at least in part by modulating macrophage function.


Sujet(s)
Athérosclérose/métabolisme , Antigènes CD70/métabolisme , Artériopathies carotidiennes/métabolisme , Macrophages/métabolisme , Plaque d'athérosclérose , Sujet âgé , Animaux , Apoptose , Athérosclérose/immunologie , Athérosclérose/anatomopathologie , Athérosclérose/prévention et contrôle , Transplantation de moelle osseuse , Antigènes CD70/déficit , Antigènes CD70/génétique , Artériopathies carotidiennes/immunologie , Artériopathies carotidiennes/anatomopathologie , Cellules cultivées , Cholestérol/métabolisme , Modèles animaux de maladie humaine , Femelle , Humains , Médiateurs de l'inflammation/métabolisme , Lipoprotéines LDL/métabolisme , Macrophages/immunologie , Macrophages/anatomopathologie , Mâle , Souris invalidées pour les gènes ApoE , Nécrose , Monoxyde d'azote/métabolisme , Phagocytose , Phénotype , Espèces réactives de l'oxygène/métabolisme , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE