Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Front Cardiovasc Med ; 10: 1171764, 2023.
Article de Anglais | MEDLINE | ID: mdl-37215541

RÉSUMÉ

Background: Atherosclerosis is the underlying cause of many cardiovascular diseases, such as myocardial infarction or stroke. B cells, and their production of pro- and anti-atherogenic antibodies, play an important role in atherosclerosis. In B cells, TRAF2 and NCK-interacting Kinase (TNIK), a germinal center kinase, was shown to bind to TNF-receptor associated factor 6 (TRAF6), and to be involved in JNK and NF-κB signaling in human B cells, a pathway associated with antibody production. Objective: We here investigate the role of TNIK-deficient B cells in atherosclerosis. Results: ApoE-/-TNIKfl/fl (TNIKBWT) and ApoE-/-TNIKfl/flCD19-cre (TNIKBKO) mice received a high cholesterol diet for 10 weeks. Atherosclerotic plaque area did not differ between TNIKBKO and TNIKBWT mice, nor was there any difference in plaque necrotic core, macrophage, T cell, α-SMA and collagen content. B1 and B2 cell numbers did not change in TNIKBKO mice, and marginal zone, follicular or germinal center B cells were unaffected. Total IgM and IgG levels, as well as oxidation specific epitope (OSE) IgM and IgG levels, did not change in absence of B cell TNIK. In contrast, plasma IgA levels were decreased in TNIKBKO mice, whereas the number of IgA+ B cells in intestinal Peyer's patches increased. No effects could be detected on T cell or myeloid cell numbers or subsets. Conclusion: We here conclude that in hyperlipidemic ApoE-/- mice, B cell specific TNIK deficiency does not affect atherosclerosis.

2.
Nat Commun ; 12(1): 3754, 2021 06 18.
Article de Anglais | MEDLINE | ID: mdl-34145241

RÉSUMÉ

Atherosclerosis is a major underlying cause of cardiovascular disease. Previous studies showed that inhibition of the co-stimulatory CD40 ligand (CD40L)-CD40 signaling axis profoundly attenuates atherosclerosis. As CD40L exerts multiple functions depending on the cell-cell interactions involved, we sought to investigate the function of the most relevant CD40L-expressing cell types in atherosclerosis: T cells and platelets. Atherosclerosis-prone mice with a CD40L-deficiency in CD4+ T cells display impaired Th1 polarization, as reflected by reduced interferon-γ production, and smaller atherosclerotic plaques containing fewer T-cells, smaller necrotic cores, an increased number of smooth muscle cells and thicker fibrous caps. Mice with a corresponding CD40-deficiency in CD11c+ dendritic cells phenocopy these findings, suggesting that the T cell-dendritic cell CD40L-CD40 axis is crucial in atherogenesis. Accordingly, sCD40L/sCD40 and interferon-γ concentrations in carotid plaques and plasma are positively correlated in patients with cerebrovascular disease. Platelet-specific deficiency of CD40L does not affect atherogenesis but ameliorates atherothrombosis. Our results establish divergent and cell-specific roles of CD40L-CD40 in atherosclerosis, which has implications for therapeutic strategies targeting this pathway.


Sujet(s)
Athérosclérose/anatomopathologie , Lymphocytes T CD4+/métabolisme , Antigènes CD40/métabolisme , Ligand de CD40/métabolisme , Interféron gamma/métabolisme , Plaque d'athérosclérose/anatomopathologie , Animaux , Plaquettes/métabolisme , Lymphocytes T CD4+/cytologie , Maladies cardiovasculaires/anatomopathologie , Cellules dendritiques/immunologie , Souris , Souris knockout , Myocytes du muscle lisse/cytologie , Transduction du signal/physiologie , Thrombose/anatomopathologie
3.
J Cardiovasc Transl Res ; 14(1): 13-22, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32222950

RÉSUMÉ

Chronic inflammation drives the development of atherosclerosis. Despite optimal treatment of classical cardiovascular risk factors, a substantial portion of the population has elevated inflammatory biomarkers and develops atherosclerosis-related complications, indicating that a residual inflammatory risk drives atherosclerotic cardiovascular disease in these patients. Additional anti-inflammatory therapeutic strategies are therefore required. The co-stimulatory molecule CD40 and its ligand CD40L (CD154) have a central role in the regulation of the inflammatory response during the development of atherosclerosis by modulating the interaction between immune cells and between immune cells and non-immune cells. In this review, we discuss the role of the CD40-CD40L dyad in atherosclerosis, and we discuss recent studies on the therapeutic potential of novel CD40-CD40L targeting strategies in cardiovascular medicine.


Sujet(s)
Antigènes CD40/immunologie , Ligand de CD40/immunologie , Maladies cardiovasculaires/étiologie , Immunothérapie/méthodes , Inflammation/complications , Animaux , Maladies cardiovasculaires/immunologie , Maladies cardiovasculaires/prévention et contrôle , Humains , Inflammation/immunologie , Inflammation/thérapie
4.
Cells ; 9(9)2020 08 29.
Article de Anglais | MEDLINE | ID: mdl-32872393

RÉSUMÉ

T cell-driven inflammation plays a critical role in the initiation and progression of atherosclerosis. The co-inhibitory protein Cytotoxic T-Lymphocyte Associated protein (CTLA) 4 is an important negative regulator of T cell activation. Here, we studied the effects of the antibody-mediated inhibition of CTLA4 on experimental atherosclerosis by treating 6-8-week-old Ldlr-/- mice, fed a 0.15% cholesterol diet for six weeks, biweekly with 200 µg of CTLA4 antibodies or isotype control for six weeks. 18F-fluorodeoxyglucose Positron Emission Tomography-Computed Tomography showed no effect of the CTLA4 inhibition of activity in the aorta, spleen, and bone marrow, indicating that monocyte/macrophage-driven inflammation was unaffected. Correspondingly, flow cytometry demonstrated that the antibody-mediated inhibition of CTLA4 did not affect the monocyte populations in the spleen. αCTLA4 treatment induced an activated T cell profile, characterized by a decrease in naïve CD44-CD62L+CD4+ T cells and an increase in CD44+CD62L- CD4+ and CD8+ T cells in the blood and lymphoid organs. Furthermore, αCTLA4 treatment induced endothelial activation, characterized by increased ICAM1 expression in the aortic endothelium. In the aortic arch, which mainly contained early atherosclerotic lesions at this time point, αCTLA4 treatment induced a 2.0-fold increase in the plaque area. These plaques had a more advanced morphological phenotype and an increased T cell/macrophage ratio, whereas the smooth muscle cell and collagen content decreased. In the aortic root, a site that contained more advanced plaques, αCTLA4 treatment increased the plaque T cell content. The short-term antibody-mediated inhibition of CTLA4 thus accelerated the progression of atherosclerosis by inducing a predominantly T cell-driven inflammation, and resulted in the formation of plaques with larger necrotic cores and less collagen. This indicates that existing therapies that are based on αCTLA4 antibodies may promote CVD development in patients.


Sujet(s)
Athérosclérose/génétique , Antigène CTLA-4/métabolisme , Hyperlipidémies/génétique , Inflammation/génétique , Animaux , Modèles animaux de maladie humaine , Évolution de la maladie , Inflammation/anatomopathologie , Mâle , Souris , Souris knockout
5.
Eur Heart J ; 41(31): 2938-2948, 2020 08 14.
Article de Anglais | MEDLINE | ID: mdl-32728688

RÉSUMÉ

AIMS: GITR-a co-stimulatory immune checkpoint protein-is known for both its activating and regulating effects on T-cells. As atherosclerosis bears features of chronic inflammation and autoimmunity, we investigated the relevance of GITR in cardiovascular disease (CVD). METHODS AND RESULTS: GITR expression was elevated in carotid endarterectomy specimens obtained from patients with cerebrovascular events (n = 100) compared to asymptomatic patients (n = 93) and correlated with parameters of plaque vulnerability, including plaque macrophage, lipid and glycophorin A content, and levels of interleukin (IL)-6, IL-12, and C-C-chemokine ligand 2. Soluble GITR levels were elevated in plasma from subjects with CVD compared to healthy controls. Plaque area in 28-week-old Gitr-/-Apoe-/- mice was reduced, and plaques had a favourable phenotype with less macrophages, a smaller necrotic core and a thicker fibrous cap. GITR deficiency did not affect the lymphoid population. RNA sequencing of Gitr-/-Apoe-/- and Apoe-/- monocytes and macrophages revealed altered pathways of cell migration, activation, and mitochondrial function. Indeed, Gitr-/-Apoe-/- monocytes displayed decreased integrin levels, reduced recruitment to endothelium, and produced less reactive oxygen species. Likewise, GITR-deficient macrophages produced less cytokines and had a reduced migratory capacity. CONCLUSION: Our data reveal a novel role for the immune checkpoint GITR in driving myeloid cell recruitment and activation in atherosclerosis, thereby inducing plaque growth and vulnerability. In humans, elevated GITR expression in carotid plaques is associated with a vulnerable plaque phenotype and adverse cerebrovascular events. GITR has the potential to become a novel therapeutic target in atherosclerosis as it reduces myeloid cell recruitment to the arterial wall and impedes atherosclerosis progression.


Sujet(s)
Athérosclérose , Protéine associée au récepteur du TNF induit par les corticoïdes , Plaque d'athérosclérose , Animaux , Apolipoprotéines E/génétique , Modèles animaux de maladie humaine , Glucocorticoïdes , Humains , Souris , Souris de lignée C57BL , Souris knockout , Phénotype , Récepteurs aux facteurs de nécrose tumorale
6.
JACC CardioOncol ; 2(4): 599-610, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-34396271

RÉSUMÉ

BACKGROUND: Immunotherapy has revolutionized cancer treatment. However, immune checkpoint inhibitors (ICIs) that target PD-1 (programmed cell death protein-1) and/or CTLA-4 (cytotoxic T lymphocyte-associated antigen-4) are commonly associated with acute immune-related adverse events. Accumulating evidence also suggests that ICIs aggravate existing inflammatory diseases. OBJECTIVES: As inflammation drives atherosclerotic cardiovascular disease, we studied the propensity of short-term ICI therapy to aggravate atherosclerosis. METHODS: We used 18F-FDG (2-deoxy-2-[fluorine-18]fluoro-D-glucose) positron emission tomography-computed tomography to detect macrophage-driven vascular and systemic inflammation in pembrolizumab and nivolumab/ipilimumab-treated melanoma patients. In parallel, atherosclerotic Ldlr -/- mice were treated with CTLA-4 and PD-1 inhibition to study the proinflammatory consequences of immune checkpoint inhibition. RESULTS: ICI treatment did not affect 18F-FDG uptake in the large arteries, spleen, and bone marrow of melanoma patients, nor myeloid cell activation in blood and lymphoid organs in hyperlipidemic mice. In contrast, we found marked changes in the adaptive immune response (i.e., increased CD4+ effector T cell and CD8+ cytotoxic T cell numbers in lymphoid organs and the arterial wall of our hyperlipidemic mice). Although plaque size was unaffected, plaques had progressed toward a lymphoid-based inflammatory phenotype, characterized by a 2.7-fold increase of CD8+ T cells and a 3.9-fold increase in necrotic core size. Increased endothelial activation was observed with a 2.2-fold and 1.6-fold increase in vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, respectively. CONCLUSIONS: This study demonstrates that combination therapy with anti-CTLA-4 and anti-PD-1 antibodies does not affect myeloid-driven vascular and systemic inflammation in melanoma patients and hyperlipidemic mice. However, short-term ICI therapy in mice induces T cell-mediated plaque inflammation and drives plaque progression.

7.
Nat Metab ; 1(9): 912-926, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31572976

RÉSUMÉ

Atherosclerosis is a progressive vascular disease triggered by interplay between abnormal shear stress and endothelial lipid retention. A combination of these and, potentially, other factors leads to a chronic inflammatory response in the vessel wall, which is thought to be responsible for disease progression characterized by a buildup of atherosclerotic plaques. Yet molecular events responsible for maintenance of plaque inflammation and plaque growth have not been fully defined. Here we show that endothelial TGFß signaling is one of the primary drivers of atherosclerosis-associated vascular inflammation. Inhibition of endothelial TGFß signaling in hyperlipidemic mice reduces vessel wall inflammation and vascular permeability and leads to arrest of disease progression and regression of established lesions. These pro-inflammatory effects of endothelial TGFß signaling are in stark contrast with its effects in other cell types and identify it as an important driver of atherosclerotic plaque growth and show the potential of cell-type specific therapeutic intervention aimed at control of this disease.


Sujet(s)
Athérosclérose/métabolisme , Endothélium vasculaire/métabolisme , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Vascularite/métabolisme , Animaux , Perméabilité capillaire , Lignée cellulaire , Évolution de la maladie , Endothélium vasculaire/anatomopathologie , Humains , Souris , Souris knockout , Facteur de croissance transformant bêta/génétique
8.
Eur Heart J ; 40(4): 372-382, 2019 01 21.
Article de Anglais | MEDLINE | ID: mdl-30452556

RÉSUMÉ

Aims: The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results: The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb-/-Apoe-/- mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb-/-Apoe-/- macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb-/-Apoe-/- CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb-/-Apoe-/- bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion: Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques.


Sujet(s)
Athérosclérose/étiologie , Lymphocytes T CD8+/immunologie , Lymphome B/complications , Macrophages/anatomopathologie , Protéine oncogène v-cbl/métabolisme , Plaque d'athérosclérose/étiologie , Animaux , Apoptose , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Modèles animaux de maladie humaine , Humains , Lymphome B/métabolisme , Lymphome B/anatomopathologie , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Phénotype , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie
9.
J Pathol ; 247(4): 471-480, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30471110

RÉSUMÉ

The costimulatory CD40L-CD40 dyad plays a major role in multiple sclerosis (MS). CD40 is highly expressed on MHCII+ B cells, dendritic cells and macrophages in human MS lesions. Here we investigated the role of the CD40 downstream signaling intermediates TNF receptor-associated factor 2 (TRAF2) and TRAF6 in MHCII+ cells in experimental autoimmune encephalomyelitis (EAE). Both MHCII-CD40-Traf2-/- and MHCII-CD40-Traf6-/- mice showed a reduction in clinical signs of EAE and prevented demyelination. However, only MHCII-CD40-Traf6-/- mice displayed a decrease in myeloid and lymphoid cell infiltration into the CNS that was accompanied by reduced levels of TNF-α, IL-6 and IFN-γ. As CD40-TRAF6 interactions predominantly occur in macrophages, we subjected CD40flfl LysMcre mice to EAE. This myeloid-specific deletion of CD40 resulted in a significant reduction in EAE severity, reduced CNS inflammation and demyelination. In conclusion, the CD40-TRAF6 signaling pathway in MHCII+ cells plays a key role in neuroinflammation and demyelination during EAE. Concomitant with the fact that CD40-TRAF6 interactions are predominant in macrophages, depletion of myeloid CD40 also reduces neuroinflammation. CD40-TRAF6 interactions thus represent a promising therapeutic target for MS. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Sujet(s)
Antigènes CD40/physiologie , Encéphalomyélite auto-immune expérimentale/immunologie , Macrophages/immunologie , Transduction du signal/immunologie , Facteur-6 associé aux récepteurs de TNF/physiologie , Animaux , Autoanticorps/métabolisme , Antigènes CD40/déficit , Ligand de CD40/physiologie , Cytokines/métabolisme , Femelle , Immunoglobuline G/immunologie , Souris de lignée C57BL , Sclérose en plaques/immunologie , Glycoprotéine MOG/immunologie , Névrite/immunologie
10.
Arterioscler Thromb Vasc Biol ; 38(5): 1076-1085, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29519940

RÉSUMÉ

OBJECTIVE: The mechanisms underlying formation of arterial aneurysms remain incompletely understood. Because inflammation is a common feature during the progressive degeneration of the aortic wall, we studied the role of the costimulatory molecule CD40L, a major driver of inflammation, in aneurysm formation. APPROACH AND RESULTS: Transcriptomics data obtained from human abdominal aortic aneurysms and normal aortas revealed increased abundance of both CD40L and CD40 in media of thrombus-free and thrombus-covered human abdominal aortic aneurysms samples. To further unravel the role of CD40L in aneurysm formation, apolipoprotein E-deficient (Apoe-/-) and Cd40l-/-Apoe-/- mice were infused with angiotensin II for 7 and 28 days. Only a minority of Cd40l-/-Apoe-/- mice (33% and 17%) developed (dissecting) aneurysms compared with 75% and 67% of Apoe-/- littermates after 7 and 28 days of infusion, respectively. Total vessel area of the aorta at the suprarenal level was 52% smaller in angiotensin II-infused Cd40l-/-Apoe-/- mice compared with that in angiotensin II-infused Apoe-/- mice. Chimeric Apoe-/- mice repopulated with Cd40l-/-Apoe-/- bone marrow afforded a similar protection against dissecting aneurysm formation. Moreover, lack of CD40L protected mice from fatal aneurysm rupture. T helper cell and macrophage accumulation in aneurysmal tissue was reduced in Cd40l-/-Apoe-/- mice with a concomitant decrease in expression of proinflammatory chemo- and cytokines. In addition, aneurysms of Cd40l-/-Apoe-/- mice displayed reduced abundance of matrix metalloproteinase-13 and an increase in tissue inhibitor of metalloproteinase-3 while activity of matrix metalloproteinase-2 and matrix metalloproteinase-9 was diminished. CONCLUSIONS: Deficiency of (hematopoietic) CD40L protects against dissecting aneurysm formation and reduces the incidence of fatal rupture. This is associated with a decreased accumulation and activation of inflammatory cells and a dampened protease activity in the arterial wall.


Sujet(s)
Aorte abdominale/métabolisme , Anévrysme de l'aorte abdominale/prévention et contrôle , 795/prévention et contrôle , Rupture aortique/prévention et contrôle , Ligand de CD40/déficit , 795/induit chimiquement , 795/génétique , 795/métabolisme , Angiotensine-II , Animaux , Aorte abdominale/anatomopathologie , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/génétique , Anévrysme de l'aorte abdominale/métabolisme , Rupture aortique/induit chimiquement , Rupture aortique/génétique , Rupture aortique/métabolisme , Ligand de CD40/génétique , Chimiokines/génétique , Chimiokines/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Dilatation pathologique , Modèles animaux de maladie humaine , Humains , Mâle , Matrix Metalloproteinase 13/génétique , Matrix Metalloproteinase 13/métabolisme , Matrix metalloproteinase 2/génétique , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 9/métabolisme , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Inhibiteur tissulaire de métalloprotéinase-3/génétique , Inhibiteur tissulaire de métalloprotéinase-3/métabolisme
11.
J Am Coll Cardiol ; 71(5): 527-542, 2018 02 06.
Article de Anglais | MEDLINE | ID: mdl-29406859

RÉSUMÉ

BACKGROUND: Disrupting the costimulatory CD40-CD40L dyad reduces atherosclerosis, but can result in immune suppression. The authors recently identified small molecule inhibitors that block the interaction between CD40 and tumor necrosis factor receptor-associated factor (TRAF) 6 (TRAF-STOPs), while leaving CD40-TRAF2/3/5 interactions intact, thereby preserving CD40-mediated immunity. OBJECTIVES: This study evaluates the potential of TRAF-STOP treatment in atherosclerosis. METHODS: The effects of TRAF-STOPs on atherosclerosis were investigated in apolipoprotein E deficient (Apoe-/-) mice. Recombinant high-density lipoprotein (rHDL) nanoparticles were used to target TRAF-STOPs to macrophages. RESULTS: TRAF-STOP treatment of young Apoe-/- mice reduced atherosclerosis by reducing CD40 and integrin expression in classical monocytes, thereby hampering monocyte recruitment. When Apoe-/- mice with established atherosclerosis were treated with TRAF-STOPs, plaque progression was halted, and plaques contained an increase in collagen, developed small necrotic cores, and contained only a few immune cells. TRAF-STOP treatment did not impair "classical" immune pathways of CD40, including T-cell proliferation and costimulation, Ig isotype switching, or germinal center formation, but reduced CD40 and ß2-integrin expression in inflammatory monocytes. In vitro testing and transcriptional profiling showed that TRAF-STOPs are effective in reducing macrophage migration and activation, which could be attributed to reduced phosphorylation of signaling intermediates of the canonical NF-κB pathway. To target TRAF-STOPs specifically to macrophages, TRAF-STOP 6877002 was incorporated into rHDL nanoparticles. Six weeks of rHDL-6877002 treatment attenuated the initiation of atherosclerosis in Apoe-/- mice. CONCLUSIONS: TRAF-STOPs can overcome the current limitations of long-term CD40 inhibition in atherosclerosis and have the potential to become a future therapeutic for atherosclerosis.


Sujet(s)
Athérosclérose/anatomopathologie , Athérosclérose/prévention et contrôle , Ligand de CD40/antagonistes et inhibiteurs , Macrophages/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Facteur-6 associé aux récepteurs de TNF/antagonistes et inhibiteurs , Dérivés de l'aniline/pharmacologie , Animaux , Techniques de culture cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Humains , Souris , Souris de lignée C57BL , Monocytes/effets des médicaments et des substances chimiques , Propiophénones/pharmacologie
12.
Cardiovasc Res ; 114(3): 368-377, 2018 03 01.
Article de Anglais | MEDLINE | ID: mdl-29309533

RÉSUMÉ

In the past decades, the inflammatory nature of atherosclerosis has been well-recognized and despite the development of therapeutic strategies targeted at its classical risk factors such as dyslipidemia and hypertension, atherosclerosis remains a major cause of morbidity and mortality. Additional strategies targeting the chronic inflammatory pathways underlying the development of atherosclerosis are therefore required. Interactions between different immune cells result in the secretion of inflammatory mediators, such as cytokines and chemokines, and fuel atherogenesis. Immune checkpoint proteins have a critical role in facilitating immune cell interactions and play an essential role in the development of atherosclerosis. Although the therapeutic potential of these molecules is well-recognized in clinical oncology, the use of immune checkpoint modulators in atherosclerosis is still limited to experimental models. Here, we review recent insights on the role of immune checkpoint proteins in atherosclerosis. Additionally, we explore the therapeutic potential and challenges of immune checkpoint modulating strategies in cardiovascular medicine and we discuss novel therapeutic approaches to target these proteins in atherosclerosis.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Artères/effets des médicaments et des substances chimiques , Athérosclérose/traitement médicamenteux , Facteurs immunologiques/usage thérapeutique , Médiateurs de l'inflammation/antagonistes et inhibiteurs , Thérapie moléculaire ciblée , Plaque d'athérosclérose , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Artères/immunologie , Artères/métabolisme , Artères/anatomopathologie , Athérosclérose/immunologie , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Conception de médicament , Humains , Médiateurs de l'inflammation/immunologie , Médiateurs de l'inflammation/métabolisme
13.
Am J Pathol ; 187(12): 2912-2919, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-28935569

RÉSUMÉ

The costimulatory molecule CD40 is a major driver of atherosclerosis. It is expressed on a wide variety of cell types, including mature dendritic cells (DCs), and is required for optimal T-cell activation and expansion. It remains undetermined whether and how CD40 on DCs impacts the pathogenesis of atherosclerosis. Here, the effects of constitutively active CD40 in DCs on atherosclerosis were examined using low-density lipoprotein-deficient (Ldlr-/-) bone marrow chimeras that express a transgene containing an engineered latent membrane protein 1 (LMP)/CD40 fusion protein conferring constitutive CD40 signaling under control of the DC-specific CD11c promoter (DC-LMP1/CD40). As expected, DC-LMP1/CD40/Ldlr-/- chimeras (DC-LMP1/CD40) showed increased antigen-presenting capacity of DCs and increased T-cell numbers. However, the mice developed extensive neutrophilia compared to CD40wt/Ldlr-/- (CD40wt) chimeras. Despite overt T-cell expansion and neutrophilia, a reduction in conventional DC frequency and a dramatic (approximately 80%) reduction in atherosclerosis was observed. Further analyses revealed that cholesterol and triglyceride levels had decreased by 37% and 60%, respectively, in DC-LMP1/CD40 chimeras. Moreover, DC-LMP1/CD40 chimeras developed inflammatory bowel disease characterized by massive transmural influx of leukocytes and lymphocytes, resulting in villous degeneration and lipid malabsorption. Constitutive activation of CD40 in DCs results in inflammation of the gastrointestinal tract, thereby impairing lipid uptake, which consequently results in attenuated atherosclerosis.


Sujet(s)
Athérosclérose/métabolisme , Antigènes CD40/métabolisme , Cholestérol/métabolisme , Cellules dendritiques/métabolisme , Transduction du signal/physiologie , Animaux , Athérosclérose/immunologie , Cellules dendritiques/immunologie , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme , Activation des lymphocytes/physiologie , Mâle , Souris , Souris de lignée C57BL , Souches mutantes de souris
14.
J Mol Endocrinol ; 59(3): 245-255, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28694301

RÉSUMÉ

Brown adipose tissue (BAT) activation and white adipose tissue (WAT) beiging can increase energy expenditure and have the potential to reduce obesity and associated diseases. The immune system is a potential target in mediating brown and beige adipocyte activation. Type 2 and anti-inflammatory immune cells contribute to metabolic homeostasis within lean WAT, with a prominent role for eosinophils and interleukin (IL)-4-induced anti-inflammatory macrophages. We determined eosinophil numbers in epididymal WAT (EpAT), subcutaneous WAT (ScAT) and BAT after 1 day, 3 days or 1 week of high-fat diet (HFD) feeding in C57Bl/6 mice. One day of HFD resulted in a rapid drop in eosinophil numbers in EpAT and BAT, and after 3 days, in ScAT. In an attempt to restore this HFD-induced drop in adipose tissue eosinophils, we treated 1-week HFD-fed mice with helminth antigens from Schistosoma mansoni or Trichuris suis and evaluated whether the well-known protective metabolic effects of helminth antigens involves BAT activation or beiging. Indeed, antigens of both helminth species induced high numbers of eosinophils in EpAT, but failed to induce beiging. In ScAT, Schistosoma mansoni antigens induced mild eosinophilia, which was accompanied by slightly more beiging. No effects were observed in BAT. To study type 2 responses on brown adipocytes directly, T37i cells were stimulated with IL-4. This increased Ucp1 expression and strongly induced the production of eosinophil chemoattractant CCL11 (+26-fold), revealing that brown adipocytes themselves can attract eosinophils. Our findings indicate that helminth antigen-induced eosinophilia fails to induce profound beiging of white adipocytes.


Sujet(s)
Tissu adipeux/immunologie , Tissu adipeux/métabolisme , Antigènes d'helminthe/immunologie , Alimentation riche en graisse , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/métabolisme , Adipocytes/métabolisme , Tissu adipeux/anatomopathologie , Tissu adipeux brun/immunologie , Tissu adipeux brun/métabolisme , Tissu adipeux brun/anatomopathologie , Animaux , Marqueurs biologiques , Chimiokine CCL11/biosynthèse , Immunité , Interleukine-4/métabolisme , Numération des leucocytes , Mâle , Souris , Protéine-1 de découplage/génétique , Protéine-1 de découplage/métabolisme
15.
J Neuroinflammation ; 14(1): 105, 2017 05 12.
Article de Anglais | MEDLINE | ID: mdl-28494768

RÉSUMÉ

BACKGROUND: The influx of leukocytes into the central nervous system (CNS) is a key hallmark of the chronic neuro-inflammatory disease multiple sclerosis (MS). Strategies that aim to inhibit leukocyte migration across the blood-brain barrier (BBB) are therefore regarded as promising therapeutic approaches to combat MS. As the CD40L-CD40 dyad signals via TNF receptor-associated factor 6 (TRAF6) in myeloid cells to induce inflammation and leukocyte trafficking, we explored the hypothesis that specific inhibition of CD40-TRAF6 interactions can ameliorate neuro-inflammation. METHODS: Human monocytes were treated with a small molecule inhibitor (SMI) of CD40-TRAF6 interactions (6877002), and migration capacity across human brain endothelial cells was measured. To test the therapeutic potential of the CD40-TRAF6-blocking SMI under neuro-inflammatory conditions in vivo, Lewis rats and C57BL/6J mice were subjected to acute experimental autoimmune encephalomyelitis (EAE) and treated with SMI 6877002 for 6 days (rats) or 3 weeks (mice). RESULTS: We here show that a SMI of CD40-TRAF6 interactions (6877002) strongly and dose-dependently reduces trans-endothelial migration of human monocytes. Moreover, upon SMI treatment, monocytes displayed a decreased production of ROS, tumor necrosis factor (TNF), and interleukin (IL)-6, whereas the production of the anti-inflammatory cytokine IL-10 was increased. Disease severity of EAE was reduced upon SMI treatment in rats, but not in mice. However, a significant reduction in monocyte-derived macrophages, but not in T cells, that had infiltrated the CNS was eminent in both models. CONCLUSIONS: Together, our results indicate that SMI-mediated inhibition of the CD40-TRAF6 pathway skews human monocytes towards anti-inflammatory cells with reduced trans-endothelial migration capacity, and is able to reduce CNS-infiltrated monocyte-derived macrophages during neuro-inflammation, but minimally ameliorates EAE disease severity. We therefore conclude that SMI-mediated inhibition of the CD40-TRAF6 pathway may represent a beneficial treatment strategy to reduce monocyte recruitment and macrophage activation in the CNS and has the potential to be used as a co-treatment to combat MS.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Antigènes CD40/métabolisme , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Monocytes/effets des médicaments et des substances chimiques , Facteur-6 associé aux récepteurs de TNF/métabolisme , Animaux , Anti-inflammatoires/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Cellules cultivées , Cervelet/métabolisme , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale/induit chimiquement , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/immunologie , Humains , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 9/métabolisme , Souris , Monocytes/immunologie , Glycoprotéine MOG/toxicité , Nitric oxide synthase type I/métabolisme , Fragments peptidiques/toxicité , Rats , Rats de lignée LEW , Espèces réactives de l'oxygène/métabolisme , Moelle spinale/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
16.
Circulation ; 136(4): 388-403, 2017 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-28450349

RÉSUMÉ

BACKGROUND: The CXCL12/CXCR4 chemokine ligand/receptor axis controls (progenitor) cell homeostasis and trafficking. So far, an atheroprotective role of CXCL12/CXCR4 has only been implied through pharmacological intervention, in particular, because the somatic deletion of the CXCR4 gene in mice is embryonically lethal. Moreover, cell-specific effects of CXCR4 in the arterial wall and underlying mechanisms remain elusive, prompting us to investigate the relevance of CXCR4 in vascular cell types for atheroprotection. METHODS: We examined the role of vascular CXCR4 in atherosclerosis and plaque composition by inducing an endothelial cell (BmxCreERT2-driven)-specific or smooth muscle cell (SMC, SmmhcCreERT2- or TaglnCre-driven)-specific deficiency of CXCR4 in an apolipoprotein E-deficient mouse model. To identify underlying mechanisms for effects of CXCR4, we studied endothelial permeability, intravital leukocyte adhesion, involvement of the Akt/WNT/ß-catenin signaling pathway and relevant phosphatases in VE-cadherin expression and function, vascular tone in aortic rings, cholesterol efflux from macrophages, and expression of SMC phenotypic markers. Finally, we analyzed associations of common genetic variants at the CXCR4 locus with the risk for coronary heart disease, along with CXCR4 transcript expression in human atherosclerotic plaques. RESULTS: The cell-specific deletion of CXCR4 in arterial endothelial cells (n=12-15) or SMCs (n=13-24) markedly increased atherosclerotic lesion formation in hyperlipidemic mice. Endothelial barrier function was promoted by CXCL12/CXCR4, which triggered Akt/WNT/ß-catenin signaling to drive VE-cadherin expression and stabilized junctional VE-cadherin complexes through associated phosphatases. Conversely, endothelial CXCR4 deficiency caused arterial leakage and inflammatory leukocyte recruitment during atherogenesis. In arterial SMCs, CXCR4 sustained normal vascular reactivity and contractile responses, whereas CXCR4 deficiency favored a synthetic phenotype, the occurrence of macrophage-like SMCs in the lesions, and impaired cholesterol efflux. Regression analyses in humans (n=259 796) identified the C-allele at rs2322864 within the CXCR4 locus to be associated with increased risk for coronary heart disease. In line, C/C risk genotype carriers showed reduced CXCR4 expression in carotid artery plaques (n=188), which was furthermore associated with symptomatic disease. CONCLUSIONS: Our data clearly establish that vascular CXCR4 limits atherosclerosis by maintaining arterial integrity, preserving endothelial barrier function, and a normal contractile SMC phenotype. Enhancing these beneficial functions of arterial CXCR4 by selective modulators might open novel therapeutic options in atherosclerosis.


Sujet(s)
Athérosclérose/métabolisme , Athérosclérose/prévention et contrôle , Cellules endothéliales/métabolisme , Récepteurs CXCR4/biosynthèse , Animaux , Athérosclérose/génétique , Perméabilité capillaire/physiologie , Femelle , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Récepteurs CXCR4/génétique
18.
FASEB J ; 30(5): 1779-88, 2016 05.
Article de Anglais | MEDLINE | ID: mdl-26813974

RÉSUMÉ

Obesity is associated with chronic low-grade inflammation, characterized by leukocytosis and inflammation in the adipose tissue. Continuous activation of the immune system is a stressor for hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Here we studied how diet-induced obesity (DIO) affects HSPC population dynamics in the BM. Eight groups of age-matched C57Bl/6 mice received a high-fat diet (45% kilocalories from fat) ranging from 1 d up to 18 wk. The obesogenic diet caused decreased proliferation of lineage(-)Sca-1(+)c-Kit(+) (LSK) cells in the BM and a general suppression of progenitor cell populations including common lymphoid progenitors and common myeloid progenitors. Within the LSK population, DIO induced a shift in stem cells that are capable of self-renewal toward maturing multipotent progenitor cells. The higher differentiation potential resulted in increased lymphoid and myeloid ex vivo colony-forming capacity. In a competitive BM transplantation, BM from obese animals showed impaired multilineage reconstitution when transplanted into chow-fed mice. Our data demonstrate that obesity stimulates the differentiation and reduces proliferation of HSPCs in the BM, leading to a decreased HSPC population. This implies that the effects of obesity on HSPCs hampers proper functioning of the immune system.-Van den Berg, S. M., Seijkens, T. T. P., Kusters, P. J. H., Beckers, L., den Toom, M., Smeets, E., Levels, J., de Winther, M. P. J., Lutgens, E. Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow.


Sujet(s)
Cellules de la moelle osseuse/physiologie , Régime alimentaire/effets indésirables , Cellules souches hématopoïétiques/physiologie , Obésité/étiologie , Animaux , Prolifération cellulaire , Souris , Souris de lignée C57BL
19.
Methods Mol Biol ; 1339: 17-40, 2015.
Article de Anglais | MEDLINE | ID: mdl-26445779

RÉSUMÉ

Atherosclerosis is an inflammatory disease of the vessel wall characterized by activation of the innate immune system, with macrophages as the main players, as well as the adaptive immune system, characterized by a Th1-dominant immune response. Cytokines play a major role in the initiation and regulation of inflammation. In recent years, many studies have investigated the role of these molecules in experimental models of atherosclerosis. While some cytokines such as TNF or IFNγ clearly had atherogenic effects, others such as IL-10 were found to be atheroprotective. However, studies investigating the different cytokines in experimental atherosclerosis revealed that the cytokine system is complex with both disease stage-dependent and site-specific effects. In this review, we strive to provide an overview of the main cytokines involved in atherosclerosis and to shed light on their individual role during atherogenesis.


Sujet(s)
Artères/immunologie , Athérosclérose/immunologie , Cytokines/immunologie , Médiateurs de l'inflammation/immunologie , Animaux , Animal génétiquement modifié , Artères/métabolisme , Artères/anatomopathologie , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Cytokines/génétique , Cytokines/métabolisme , Modèles animaux de maladie humaine , Prédisposition génétique à une maladie , Humains , Médiateurs de l'inflammation/métabolisme , Phénotype , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE