Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 379
Filter
Add more filters

Publication year range
1.
Circ Res ; 135(4): 488-502, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38979610

ABSTRACT

BACKGROUND: The long isoform of the Wnk1 (with-no-lysine [K] kinase 1) is a ubiquitous serine/threonine kinase, but its role in vascular smooth muscle cells (VSMCs) pathophysiology remains unknown. METHODS: AngII (angiotensin II) was infused in Apoe-/- to induce experimental aortic aneurysm. Mice carrying an Sm22-Cre allele were cross-bred with mice carrying a floxed Wnk1 allele to specifically investigate the functional role of Wnk1 in VSMCs. RESULTS: Single-cell RNA-sequencing of the aneurysmal abdominal aorta from AngII-infused Apoe-/- mice revealed that VSMCs that did not express Wnk1 showed lower expression of contractile phenotype markers and increased inflammatory activity. Interestingly, WNK1 gene expression in VSMCs was decreased in human abdominal aortic aneurysm. Wnk1-deficient VSMCs lost their contractile function and exhibited a proinflammatory phenotype, characterized by the production of matrix metalloproteases, as well as cytokines and chemokines, which contributed to local accumulation of inflammatory macrophages, Ly6Chi monocytes, and γδ T cells. Sm22Cre+Wnk1lox/lox mice spontaneously developed aortitis in the infrarenal abdominal aorta, which extended to the thoracic area over time without any negative effect on long-term survival. AngII infusion in Sm22Cre+Wnk1lox/lox mice aggravated the aortic disease, with the formation of lethal abdominal aortic aneurysms. Pharmacological blockade of γδ T-cell recruitment using neutralizing anti-CXCL9 (anti-CXC motif chemokine ligand 9) antibody treatment, or of monocyte/macrophage using Ki20227, a selective inhibitor of CSF1 receptor, attenuated aortitis. Wnk1 deletion in VSMCs led to aortic wall remodeling with destruction of elastin layers, increased collagen content, and enhanced local TGF-ß (transforming growth factor-beta) 1 expression. Finally, in vivo TGF-ß blockade using neutralizing anti-TGF-ß antibody promoted saccular aneurysm formation and aorta rupture in Sm22 Cre+ Wnk1lox/lox mice but not in control animals. CONCLUSION: Wnk1 is a key regulator of VSMC function. Wnk1 deletion promotes VSMC phenotype switch toward a pathogenic proinflammatory phenotype, orchestrating deleterious vascular remodeling and spontaneous severe aortitis in mice.


Subject(s)
Angiotensin II , Aortic Aneurysm, Abdominal , Aortitis , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , WNK Lysine-Deficient Protein Kinase 1 , Animals , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Humans , WNK Lysine-Deficient Protein Kinase 1/genetics , WNK Lysine-Deficient Protein Kinase 1/metabolism , Mice, Inbred C57BL , Male , Cells, Cultured , Mice, Knockout, ApoE , Disease Models, Animal , Inflammation/metabolism , Inflammation/genetics , Inflammation/pathology , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology
2.
Rheumatology (Oxford) ; 63(9): 2473-2483, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38648749

ABSTRACT

OBJECTIVES: Epigenetically modified fibroblasts contribute to chronicity in inflammatory diseases. Reasons for the relapsing character of large vessel vasculitis (LVV) remain obscure, including the role of fibroblasts, in part due to limited access to biopsies of involved tissue.68Ga FAPI-46 (FAPI)-PET/CT detects activated fibroblasts in vivo. In this exploratory pilot study, we tested the detection of fibroblast activation in vessel walls using FAPI-PET/CT in LVV with aortitis. METHODS: Eight LVV patients with aortitis and eight age- and gender-matched controls were included. The distribution of FAPI uptake was evaluated in the aorta and large vessels. FAPI-uptake was compared with MRI inflammatory activity scores. Imaging results were compared with clinical parameters such as serum inflammatory markers, time of remission and medication. RESULTS: Three aortitis patients were clinically active and five in remission. Irrespective of activity, FAPI uptake was significantly enhanced in aortitis compared with controls. Patients in remission had a mean duration of remission of 2.8 years (range 1-4 years), yet significant FAPI uptake in the vessel wall was found. In remitted aortitis, MRI inflammatory scores were close to be negative, while in 4/5 patients visually identifiable FAPI uptake was observed. CONCLUSIONS: This pilot feasibility study shows significant tracer uptake in the aortic walls in LVV. FAPI positivity indicates ongoing fibroblast pathology in clinically remitted LVV.


Subject(s)
Aortitis , Fibroblasts , Positron Emission Tomography Computed Tomography , Humans , Male , Female , Aortitis/diagnostic imaging , Aortitis/pathology , Middle Aged , Positron Emission Tomography Computed Tomography/methods , Fibroblasts/metabolism , Fibroblasts/pathology , Pilot Projects , Aged , Aorta/diagnostic imaging , Aorta/pathology , Case-Control Studies , Gallium Radioisotopes , Endopeptidases , Adult , Magnetic Resonance Imaging/methods , Remission Induction , Membrane Proteins
3.
Scand J Rheumatol ; 52(3): 306-316, 2023 05.
Article in English | MEDLINE | ID: mdl-36763458

ABSTRACT

Aortic involvement in immunoglobulin G4-related disease (IgG4-RD) is extremely rare and is often overlooked during the aortitis work-up. IgG4-related aortitis differs from non-IgG4-related aortitis in its histopathological features, site of involvement, laboratory markers, and treatment options. The histopathological examination of the vessel walls characteristically reveals adventitial thickening with intimal sparing, typically affecting the infrarenal abdominal aorta. In addition, inadequate knowledge about the disease often leads to delayed or missed diagnosis and undermanagement of a potentially treatable condition. Hence, in this paper, we review the unique clinical manifestations, laboratory markers, diagnostic features, current treatment strategies, and novel experimental therapeutic options in the management of IgG4-related aortitis.


Subject(s)
Aortitis , Immunoglobulin G4-Related Disease , Humans , Aortitis/diagnosis , Aortitis/pathology , Immunoglobulin G4-Related Disease/diagnosis , Immunoglobulin G , Biomarkers
4.
Circ Res ; 126(1): 25-37, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31647755

ABSTRACT

RATIONALE: Through localized delivery of rapamycin via a biomimetic drug delivery system, it is possible to reduce vascular inflammation and thus the progression of vascular disease. OBJECTIVE: Use biomimetic nanoparticles to deliver rapamycin to the vessel wall to reduce inflammation in an in vivo model of atherosclerosis after a short dosing schedule. METHODS AND RESULTS: Biomimetic nanoparticles (leukosomes) were synthesized using membrane proteins purified from activated J774 macrophages. Rapamycin-loaded nanoparticles were characterized using dynamic light scattering and were found to have a diameter of 108±2.3 nm, a surface charge of -15.4±14.4 mV, and a polydispersity index of 0.11 +/ 0.2. For in vivo studies, ApoE-/- mice were fed a high-fat diet for 12 weeks. Mice were injected with either PBS, free rapamycin (5 mg/kg), or rapamycin-loaded leukosomes (Leuko-Rapa; 5 mg/kg) once daily for 7 days. In mice treated with Leuko-Rapa, flow cytometry of disaggregated aortic tissue revealed fewer proliferating macrophages in the aorta (15.6±9.79 %) compared with untreated mice (30.2±13.34 %) and rapamycin alone (26.8±9.87 %). Decreased macrophage proliferation correlated with decreased levels of MCP (monocyte chemoattractant protein)-1 and IL (interleukin)-b1 in mice treated with Leuko-Rapa. Furthermore, Leuko-Rapa-treated mice also displayed significantly decreased MMP (matrix metalloproteinases) activity in the aorta (mean difference 2554±363.9, P=9.95122×10-6). No significant changes in metabolic or inflammation markers observed in liver metabolic assays. Histological analysis showed improvements in lung morphology, with no alterations in heart, spleen, lung, or liver in Leuko-Rapa-treated mice. CONCLUSIONS: We showed that our biomimetic nanoparticles showed a decrease in proliferating macrophage population that was accompanied by the reduction of key proinflammatory cytokines and changes in plaque morphology. This proof-of-concept showed that our platform was capable of suppressing macrophage proliferation within the aorta after a short dosing schedule (7 days) and with a favorable toxicity profile. This treatment could be a promising intervention for the acute stabilization of late-stage plaques.


Subject(s)
Aortitis/drug therapy , Atherosclerosis/drug therapy , Mechanistic Target of Rapamycin Complex 1/drug effects , Plaque, Atherosclerotic/prevention & control , Sirolimus/administration & dosage , 1,2-Dipalmitoylphosphatidylcholine/administration & dosage , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortitis/complications , Aortitis/pathology , Apolipoproteins E/deficiency , Atherosclerosis/pathology , Biomimetics , C-Reactive Protein/metabolism , Cryoelectron Microscopy , Cytokines/metabolism , Drug Evaluation, Preclinical , Macrophage Activation/drug effects , Macrophages/metabolism , Membrane Proteins/administration & dosage , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Neovascularization, Pathologic/prevention & control , Organ Specificity , Phosphatidylcholines/administration & dosage , Random Allocation , Sirolimus/pharmacology , Sirolimus/therapeutic use
5.
Arterioscler Thromb Vasc Biol ; 41(1): 331-345, 2021 01.
Article in English | MEDLINE | ID: mdl-33147994

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a vascular degenerative disease causing sudden rupture of aorta and significant mortality in elders. Nevertheless, no prognostic and therapeutic target is available for disease management. Gal-1 (galectin-1) is a ß-galactoside-binding lectin constitutively expressed in vasculature with roles in maintaining vascular homeostasis. This study aims to investigate the potential involvement of Gal-1 in AAA progression. Approach and Results: Gal-1 was significantly elevated in circulation and aortic tissues of Ang II (angiotensin II)-infused apoE-deficient mice developing AAA. Gal-1 deficiency reduced incidence and severity of AAA with lower expression of aortic MMPs (matrix metalloproteases) and proinflammatory cytokines. TNFα (tumor necrosis factor alpha) induced Gal-1 expression in cultured vascular smooth muscle cells and adventitial fibroblasts. Gal-1 deletion enhanced TNFα-induced MMP9 expression in fibroblasts but not vascular smooth muscle cells. Cysteinyl-labeling assay demonstrated that aortic Gal-1 exhibited susceptibility to oxidation in vivo. Recombinant oxidized Gal-1 induced expression of MMP9 and inflammatory cytokines to various extents in macrophages, vascular smooth muscle cells, and fibroblasts through activation of MAP (mitogen-activated protein) kinase signaling. Clinically, serum MMP9 level was significantly higher in both patients with AAA and coronary artery disease than in control subjects, whereas serum Gal-1 level was elevated in patients with AAA but not coronary artery disease when compared with controls. CONCLUSIONS: Gal-1 is highly induced and contributes to AAA by enhancing matrix degradation activity and inflammatory responses in experimental model. The pathological link between Gal-1 and AAA is also observed in human patients. These findings support the potential of Gal-1 as a disease biomarker and therapeutic target of AAA.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Aortitis/metabolism , Galectin 1/metabolism , Vascular Remodeling , Adventitia/metabolism , Adventitia/pathology , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Aortitis/chemically induced , Aortitis/pathology , Case-Control Studies , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Galectin 1/blood , Galectin 1/deficiency , Galectin 1/genetics , Humans , Inflammation Mediators/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Signal Transduction , Up-Regulation
6.
J Cell Physiol ; 236(6): 4555-4564, 2021 06.
Article in English | MEDLINE | ID: mdl-33241567

ABSTRACT

Selenium (Se) is an essential trace element in organism. Se deficiency can cause many diseases, including vascular disease. Studies have shown that inflammation is the main inducement of vascular disease, microRNA (miRNA) can influence inflammation in various ways, and Se deficiency can affect miRNAs expression. To study the mechanism of aorta damage caused by Se deficiency, we constructed a Se deficiency porcine aorta model and found that Se deficiency can significantly inhibit miR-223, which downregulates the expression of nucleotide-binding oligomerization domain-like receptor family 3 (NLRP3). Subsequently, we found that in Se deficiency group, NLRP3, and its downstream (caspase-1, apoptosis-related spot-like protein [ASC], IL-18, IL-1ß) expression was significantly increased. In vitro, we cultured pig iliac endothelium cell lines, and constructed miR-223 knockdown and overexpression models. NLRP3 messenger RNA and protein levels were significant increased in the knockdown group, and decreased in the overexpression group. The results of this study show that Se deficiency in porcine arteries can induce inflammation through miR-223/NLRP3.


Subject(s)
Aorta/metabolism , Aortitis/metabolism , Endothelial Cells/metabolism , Inflammasomes/metabolism , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Selenium/deficiency , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Aorta/immunology , Aorta/pathology , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/pathology , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , MicroRNAs/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Sus scrofa
7.
Am J Physiol Heart Circ Physiol ; 320(5): H1836-H1850, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33666505

ABSTRACT

Fine particulate matter (PM2.5) air pollution exposure increases the risk of developing cardiovascular disease (CVD). Although the precise mechanisms by which air pollution exposure increases CVD risk remain uncertain, research indicates that PM2.5-induced endothelial dysfunction contributes to CVD risk. Previous studies demonstrate that concentrated ambient PM2.5 (CAP) exposure induces vascular inflammation and impairs insulin and vascular endothelial growth factor (VEGF) signaling dependent on pulmonary oxidative stress. To assess whether CAP exposure induces these vascular effects via plasmatic factors, we incubated aortas from naïve mice with plasma isolated from mice exposed to HEPA-filtered air or CAP (9 days) and examined vascular inflammation and insulin and VEGF signaling. We found that treatment of naïve aortas with plasma from CAP-exposed mice activates NF-κBα and induces insulin and VEGF resistance, indicating transmission by plasmatic factor(s). To identify putative factors, we exposed lung-specific ecSOD-transgenic (ecSOD-Tg) mice and wild-type (WT) littermates to CAP at concentrations of either ∼60 µg/m3 (CAP60) or ∼100 µg/m3 (CAP100) and measured the abundance of plasma metabolites by mass spectrometry. In WT mice, both CAP concentrations increased levels of fatty acids such as palmitate, myristate, and palmitoleate and decreased numerous phospholipid species; however, these CAP-induced changes in the plasma lipidome were prevented in ecSOD-Tg mice. Consistent with the literature, we found that fatty acids such as palmitate are sufficient to promote endothelial inflammation. Collectively, our findings suggest that PM2.5 exposure, by inducing pulmonary oxidative stress, promotes unique lipidomic changes characterized by high levels of circulating fatty acids, which are sufficient to trigger vascular pathology.NEW & NOTEWORTHY We found that circulating plasma constituents are responsible for air pollution-induced vascular pathologies. Inhalation of fine particulate matter (≤PM2.5) promotes a unique form of dyslipidemia that manifests in a manner dependent upon pulmonary oxidative stress. The air pollution-engendered dyslipidemic phenotype is characterized by elevated free fatty acid species and diminished phospholipid species, which could contribute to vascular inflammation and loss of insulin sensitivity.


Subject(s)
Air Pollutants/toxicity , Aorta/drug effects , Aortitis/chemically induced , Dyslipidemias/chemically induced , Insulin Resistance , Lipids/blood , Metabolome , Particulate Matter/toxicity , Animals , Aorta/metabolism , Aorta/pathology , Aortitis/blood , Aortitis/pathology , Biomarkers/blood , Cells, Cultured , Dyslipidemias/blood , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inhalation Exposure , Insulin/blood , Lipidomics , Lung/drug effects , Lung/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/drug effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/blood
8.
Arterioscler Thromb Vasc Biol ; 40(9): 2159-2170, 2020 09.
Article in English | MEDLINE | ID: mdl-32673527

ABSTRACT

OBJECTIVE: Evidence from preclinical and clinical studies has demonstrated that myocardial infarction promotes atherosclerosis progression. The impact of focal vascular inflammation on the progression and phenotype of remote atherosclerosis remains unknown. Approach and Results: We used a novel ApoE-/- knockout mouse model of sustained arterial inflammation, initiated by mechanical injury in the abdominal aorta. Using serial in vivo molecular MRI and ex vivo histology and flow cytometry, we demonstrate that focal arterial inflammation triggered by aortic injury, accelerates atherosclerosis in the remote brachiocephalic artery. The brachiocephalic artery atheroma had distinct histological features including increased plaque size, plaque permeability, necrotic core to collagen ratio, infiltration of more inflammatory monocyte subsets, and reduced collagen content. We also found that arterial inflammation following focal vascular injury evoked a prolonged systemic inflammatory response manifested as a persistent increase in serum IL-6 (interleukin 6). Finally, we demonstrate that 2 therapeutic interventions-pravastatin and minocycline-had distinct anti-inflammatory effects at the plaque and systemic level. CONCLUSIONS: We show for the first time that focal arterial inflammation in response to vascular injury enhances systemic vascular inflammation, accelerates remote atheroma progression and induces plaques more inflamed, lipid-rich, and collagen-poor in the absence of ischemic myocardial injury. This inflammatory cascade is modulated by pravastatin and minocycline treatments, which have anti-inflammatory effects at both plaque and systemic levels that mitigate atheroma progression.


Subject(s)
Aortitis/complications , Atherosclerosis/etiology , Brachiocephalic Trunk/metabolism , Inflammation Mediators/blood , Plaque, Atherosclerotic , Animals , Anti-Inflammatory Agents/pharmacology , Aortitis/blood , Aortitis/pathology , Atherosclerosis/blood , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/pathology , Collagen/metabolism , Disease Models, Animal , Disease Progression , Interleukin-6/blood , Lipid Metabolism , Male , Mice, Knockout, ApoE , Minocycline/pharmacology , Necrosis , Pravastatin/pharmacology , Time Factors
9.
Arterioscler Thromb Vasc Biol ; 40(10): 2494-2507, 2020 10.
Article in English | MEDLINE | ID: mdl-32787523

ABSTRACT

OBJECTIVE: Currently, there are no approved drugs for abdominal aortic aneurysm (AAA) treatment, likely due to limited understanding of the primary molecular mechanisms underlying AAA development and progression. BAF60a-a unique subunit of the SWI/SNF (switch/sucrose nonfermentable) chromatin remodeling complex-is a novel regulator of metabolic homeostasis, yet little is known about its function in the vasculature and pathogenesis of AAA. In this study, we sought to investigate the role and underlying mechanisms of vascular smooth muscle cell (VSMC)-specific BAF60a in AAA formation. Approach and Results: BAF60a is upregulated in human and experimental murine AAA lesions. In vivo studies revealed that VSMC-specific knockout of BAF60a protected mice from both Ang II (angiotensin II)-induced and elastase-induced AAA formation with significant suppression of vascular inflammation, monocyte infiltration, and elastin fragmentation. Through RNA sequencing and pathway analysis, we found that the expression of inflammatory response genes in cultured human aortic smooth muscle cells was significantly downregulated by small interfering RNA-mediated BAF60a knockdown while upregulated upon adenovirus-mediated BAF60a overexpression. BAF60a regulates VSMC inflammation by recruiting BRG1 (Brahma-related gene-1)-a catalytic subunit of the SWI/SNF complex-to the promoter region of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) target genes. Furthermore, loss of BAF60a in VSMCs prevented the upregulation of the proteolytic enzyme cysteine protease CTSS (cathepsin S), thus ameliorating ECM (extracellular matrix) degradation within the vascular wall in AAA. CONCLUSIONS: Our study demonstrated that BAF60a is required to recruit the SWI/SNF complex to facilitate the epigenetic regulation of VSMC inflammation, which may serve as a potential therapeutic target in preventing and treating AAA.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Aortitis/prevention & control , Chromosomal Proteins, Non-Histone/deficiency , Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Vascular Remodeling , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Case-Control Studies , Cathepsins/metabolism , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Extracellular Matrix/pathology , Humans , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Signal Transduction
10.
Cardiol Young ; 31(1): 132-134, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33040742
11.
Circulation ; 139(10): 1320-1334, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30586743

ABSTRACT

BACKGROUND: The majority of the human genome comprises noncoding sequences, which are in part transcribed as long noncoding RNAs (lncRNAs). lncRNAs exhibit multiple functions, including the epigenetic control of gene expression. In this study, the effect of the lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) on atherosclerosis was examined. METHODS: The effect of MALAT1 on atherosclerosis was determined in apolipoprotein E-deficient (Apoe-/-) MALAT1-deficient (Malat1-/-) mice that were fed with a high-fat diet and by studying the regulation of MALAT1 in human plaques. RESULTS: Apoe-/- Malat1-/- mice that were fed a high-fat diet showed increased plaque size and infiltration of inflammatory CD45+ cells compared with Apoe-/- Malat1+/+ control mice. Bone marrow transplantation of Apoe-/- Malat1-/- bone marrow cells in Apoe-/- Malat1+/+ mice enhanced atherosclerotic lesion formation, which suggests that hematopoietic cells mediate the proatherosclerotic phenotype. Indeed, bone marrow cells isolated from Malat1-/- mice showed increased adhesion to endothelial cells and elevated levels of proinflammatory mediators. Moreover, myeloid cells of Malat1-/- mice displayed enhanced adhesion to atherosclerotic arteries in vivo. The anti-inflammatory effects of MALAT1 were attributed in part to reduction of the microRNA miR-503. MALAT1 expression was further significantly decreased in human plaques compared with normal arteries and was lower in symptomatic versus asymptomatic patients. Lower levels of MALAT1 in human plaques were associated with a worse prognosis. CONCLUSIONS: Reduced levels of MALAT1 augment atherosclerotic lesion formation in mice and are associated with human atherosclerotic disease. The proatherosclerotic effects observed in Malat1-/- mice were mainly caused by enhanced accumulation of hematopoietic cells.


Subject(s)
Aorta/metabolism , Aortitis/metabolism , Atherosclerosis/metabolism , Bone Marrow Cells/metabolism , Hematopoiesis , Plaque, Atherosclerotic , RNA, Long Noncoding/metabolism , Animals , Aorta/pathology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Case-Control Studies , Disease Models, Animal , Down-Regulation , Humans , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Signal Transduction
12.
Ann Rheum Dis ; 79(1): 19-30, 2020 01.
Article in English | MEDLINE | ID: mdl-31270110

ABSTRACT

BACKGROUND: Since the publication of the European League Against Rheumatism (EULAR) recommendations for the management of large vessel vasculitis (LVV) in 2009, several relevant randomised clinical trials and cohort analyses have been published, which have the potential to change clinical care and therefore supporting the need to update the original recommendations. METHODS: Using EULAR standardised operating procedures for EULAR-endorsed recommendations, the EULAR task force undertook a systematic literature review and sought opinion from 20 experts from 13 countries. We modified existing recommendations and created new recommendations. RESULTS: Three overarching principles and 10 recommendations were formulated. We recommend that a suspected diagnosis of LVV should be confirmed by imaging or histology. High dose glucocorticoid therapy (40-60 mg/day prednisone-equivalent) should be initiated immediately for induction of remission in active giant cell arteritis (GCA) or Takayasu arteritis (TAK). We recommend adjunctive therapy in selected patients with GCA (refractory or relapsing disease, presence of an increased risk for glucocorticoid-related adverse events or complications) using tocilizumab. Methotrexate may be used as an alternative. Non-biological glucocorticoid-sparing agents should be given in combination with glucocorticoids in all patients with TAK and biological agents may be used in refractory or relapsing patients. We no longer recommend the routine use of antiplatelet or anticoagulant therapy for treatment of LVV unless it is indicated for other reasons. CONCLUSIONS: We have updated the recommendations for the management of LVV to facilitate the translation of current scientific evidence and expert opinion into better management and improved outcome of patients in clinical practice.


Subject(s)
Antirheumatic Agents/therapeutic use , Giant Cell Arteritis/drug therapy , Glucocorticoids/therapeutic use , Takayasu Arteritis/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Aortitis/diagnostic imaging , Aortitis/drug therapy , Aortitis/pathology , Giant Cell Arteritis/diagnostic imaging , Giant Cell Arteritis/pathology , Humans , Methotrexate/therapeutic use , Takayasu Arteritis/diagnostic imaging , Takayasu Arteritis/pathology
13.
Clin Sci (Lond) ; 134(20): 2771-2787, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33064141

ABSTRACT

The aim of the present study was to examine whether inhibition of Interleukin (IL)-6 signaling by MR16-1, an IL-6 receptor antibody, attenuates aortitis, cardiac hypertrophy, and arthritis in IL-1 receptor antagonist deficient (IL-1RA KO) mice. Four weeks old mice were intraperitoneally administered with either MR16-1 or non-immune IgG at dosages that were adjusted over time for 5 weeks. These mice were stratified into four groups: MR16-1 treatment groups, KO/MR low group (first 2.0 mg, following 0.5 mg/week, n=14) and KO/MR high group (first 4.0 mg, following 2.0 mg/week, n=19) in IL-1RA KO mice, and IgG treatment groups, KO/IgG group (first 2.0 mg, following 1.0 mg/week, n=22) in IL-1RA KO mice, and wild/IgG group (first 2.0 mg, following 1.0 mg/week, n=17) in wild mice. Aortitis, cardiac hypertrophy and arthropathy were histologically analyzed. Sixty-eight percent of the KO/IgG group developed aortitis (53% developed severe aortitis). In contrast, only 21% of the KO/MR high group developed mild aortitis, without severe aortitis (P<0.01, vs KO/IgG group). Infiltration of inflammatory cells, such as neutrophils, T cells, and macrophages, was frequently observed around aortic sinus of the KO/IgG group. Left ventricle and cardiomyocyte hypertrophy were observed in IL-1RA KO mice. Administration of high dosage of MR16-1 significantly suppressed cardiomyocyte hypertrophy. MR16-1 attenuated the incidence and severity of arthritis in IL-1RA KO mice in a dose-dependent manner. In conclusion, blockade of IL-6 signaling may exert a beneficial effect to attenuate severe aortitis, left ventricle hypertrophy, and arthritis.


Subject(s)
Aortitis/metabolism , Arthritis/metabolism , Hypertrophy, Left Ventricular/metabolism , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin-6/metabolism , Signal Transduction , Animals , Antibodies/pharmacology , Aortitis/pathology , Arteries/pathology , Body Weight , Female , Hemodynamics , Immunity, Innate , Inflammation/pathology , Interleukin 1 Receptor Antagonist Protein/metabolism , Male , Mice, Knockout , Organ Size , Sinus of Valsalva/pathology
14.
Rheumatol Int ; 40(10): 1725-1732, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32206878

ABSTRACT

IgG4-related disease (IgG4-RD) is a fibro-inflammatory condition characterized by increased serum IgG4 level, infiltration of lymphocytes and IgG4-positive (IgG4+) plasma cells and fibrosis. It can occur in almost all organs, commonly affecting the pancreas, biliary tract, salivary and lacrimal glands and kidneys. However, reports of IgG4-RD accompanied by pathologically confirmed, IgG4-related pleural disease are scarce. Here, we present a case of a 64-year-old man with suspected malignant pleural mesothelioma based on imaging findings but finally diagnosed with IgG4-RD (including pleuritis, periaortitis and bilateral submandibular gland enlargement) based on a high serum IgG4 level and pleural histopathological findings such as lymphoplasmacytic infiltration including IgG4+ plasma cells and fibrosis. Systemic corticosteroid therapy was effective at reducing serum IgG4, improving bilateral submandibular gland enlargement, and regressing pleural thickening and periaortic soft tissue. We also discuss clinical characteristics and pleural pathological features of previously reported cases with IgG4-related pleural disease based on a comprehensive literature review. Our case of IgG4-RD with pleura, aorta and submandibular gland involvement, pathologically confirmed by pleural specimen might be unique and very rare.


Subject(s)
Aortitis/pathology , Immunoglobulin G4-Related Disease/diagnosis , Submandibular Gland/pathology , Adult , Aged , Aortitis/diagnosis , Aortitis/drug therapy , Aortitis/etiology , Diagnosis, Differential , Female , Glucocorticoids/administration & dosage , Humans , Immunoglobulin G4-Related Disease/drug therapy , Immunoglobulin G4-Related Disease/pathology , Male , Mesothelioma, Malignant/diagnosis , Middle Aged , Pleura/pathology , Prednisolone/administration & dosage
15.
Int J Mol Sci ; 21(8)2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32331251

ABSTRACT

Atherosclerosis represents one of the main risk factors for the development of cardiovascular diseases. Their etiologies have been studied in recent years in order to better define therapeutic targets for intervention and to identify diagnostic methods. Two different subtypes of macrophages, M1 and M2, have been described in physiological conditions. They can also be found in the atherosclerotic process, where they both have opposite roles in disease progression. Perivascular brown adipose tissue is also involved in inflammation and endothelial damage. In this work, we provide insights into the protective role of melatonin in the atherosclerotic process by morphological and 18F-FDG-PET/CT analyses. In particular, we examined the effects of melatonin on pathways that are linked to atherosclerosis development. We showed that melatonin, by suppressing M1 activity, reduced inflammation and directed macrophage polarization toward the M2 macrophage subtype. Moreover, melatonin preserved the activity of perivascular brown adipose tissue. In addition, 18F-FDG uptake is very high in mice treated with melatonin, confirming that other factors may alter 18F-FDG distribution. In conclusion, we showed that melatonin affects inflammatory pathways that have been linked to atherosclerosis, assessed the relationships of the 18F-FDG PET/CT parameters with macrophage markers and the production of their cytokines, which that have been defined by morphological evaluations.


Subject(s)
Apolipoproteins E/deficiency , Fluorodeoxyglucose F18 , Melatonin/metabolism , Molecular Imaging , Positron Emission Tomography Computed Tomography , Adipose Tissue, Brown/metabolism , Animals , Aortitis/etiology , Aortitis/metabolism , Aortitis/pathology , Atherosclerosis/diagnostic imaging , Atherosclerosis/etiology , Atherosclerosis/metabolism , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Molecular Imaging/methods , Radiopharmaceuticals
16.
Circulation ; 138(16): 1706-1719, 2018 10 16.
Article in English | MEDLINE | ID: mdl-29700120

ABSTRACT

BACKGROUND: The coagulation system is closely linked with vascular inflammation, although the underlying mechanisms are still obscure. Recent studies show that protease-activated receptor (PAR)-2, a major receptor of activated factor X, is expressed in both vascular cells and leukocytes, suggesting that PAR-2 may contribute to the pathogenesis of inflammatory diseases. Here we investigated the role of PAR-2 in vascular inflammation and atherogenesis. METHODS: We generated apolipoprotein E-deficient ( ApoE-/-) mice lacking systemic PAR-2 expression ( PAR-2-/- ApoE-/-). ApoE-/- mice, which lack or express PAR-2 only in bone marrow (BM) cells, were also generated by BM transplantation. Atherosclerotic lesions were investigated after 20 weeks on a Western-type diet by histological analyses, quantitative reverse transcription polymerase chain reaction, and Western blotting. In vitro experiments using BM-derived macrophages were performed to confirm the proinflammatory roles of PAR-2. The association between plasma activated factor X level and the severity of coronary atherosclerosis was also examined in humans who underwent coronary intervention. RESULTS: PAR-2-/- ApoE-/- mice showed reduced atherosclerotic lesions in the aortic arch ( P<0.05) along with features of stabilized atherosclerotic plaques, such as less lipid deposition ( P<0.05), collagen loss ( P<0.01), macrophage accumulation ( P<0.05), and inflammatory molecule expression ( P<0.05) compared with ApoE-/- mice. Systemic PAR2 deletion in ApoE-/-mice significantly decreased the expression of inflammatory molecules in the aorta. The results of BM transplantation experiments demonstrated that PAR-2 in hematopoietic cells contributed to atherogenesis in ApoE-/- mice. PAR-2 deletion did not alter metabolic parameters. In vitro experiments demonstrated that activated factor X or a specific peptide agonist of PAR-2 significantly increased the expression of inflammatory molecules and lipid uptake in BM-derived macrophages from wild-type mice compared with those from PAR-2-deficient mice. Activation of nuclear factor-κB signaling was involved in PAR-2-associated vascular inflammation and macrophage activation. In humans who underwent coronary intervention, plasma activated factor X level independently correlated with the severity of coronary atherosclerosis as determined by Gensini score ( P<0.05) and plaque volume ( P<0.01). CONCLUSIONS: PAR-2 signaling activates macrophages and promotes vascular inflammation, increasing atherosclerosis in ApoE-/- mice. This signaling pathway may also participate in atherogenesis in humans.


Subject(s)
Aorta, Thoracic/metabolism , Aortitis/metabolism , Atherosclerosis/metabolism , Inflammation Mediators/metabolism , Macrophage Activation , Macrophages/metabolism , Plaque, Atherosclerotic , Receptor, PAR-2/metabolism , Aged , Animals , Aorta, Thoracic/pathology , Aortitis/genetics , Aortitis/pathology , Aortitis/prevention & control , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Diet, Western , Disease Models, Animal , Factor Xa/metabolism , Female , Humans , Lipids/blood , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
17.
Scand J Rheumatol ; 48(6): 474-481, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31766965

ABSTRACT

Objective: To compare the clinical presentation and outcome of giant cell arteritis (GCA)-related aortitis according to the results of temporal artery biopsy (TAB).Method: Patients with GCA-related aortitis diagnosed between 2000 and 2017, who underwent TAB, were retrospectively included from a French multicentre database. They all met at least three American College of Rheumatology criteria for the diagnosis of GCA. Aortitis was defined by aortic wall thickening > 2 mm on computed tomography scan and/or an aortic aneurysm, associated with an inflammatory syndrome. Patients were divided into two groups [positive and negative TAB (TAB+, TAB-)], which were compared regarding aortic imaging characteristics and aortic events, at aortitis diagnosis and during follow-up.Results: We included 56 patients with TAB+ (70%) and 24 with TAB- (30%). At aortitis diagnosis, patients with TAB- were significantly younger than those with TAB+ (67.7 ± 9 vs 72.3 ± 7 years, p = 0.022). Initial clinical signs of GCA, inflammatory parameters, and glucocorticoid therapy were similar in both groups. Coronary artery disease and/or lower limb peripheral arterial disease was more frequent in TAB- patients (25% vs 5.3%, p = 0.018). Aortic wall thickness and type of aortic involvement were not significantly different between groups. Diffuse arterial involvement from the aortic arch was more frequent in TAB- patients (29.1 vs 8.9%, p = 0.03). There were no differences between the groups regarding overall, aneurism-free, relapse-free, and aortic event-free survival.Conclusion: Among patients with GCA-related aortitis, those with TAB- are characterized by younger age and increased frequency of diffuse arterial involvement from the aortic arch compared to those with TAB+, without significant differences in terms of prognosis.


Subject(s)
Aortitis/pathology , Giant Cell Arteritis/pathology , Temporal Arteries/pathology , Aged , Aortitis/diagnostic imaging , Aortitis/mortality , Biopsy , Female , Giant Cell Arteritis/diagnostic imaging , Giant Cell Arteritis/mortality , Humans , Male , Middle Aged , Retrospective Studies , Tomography, X-Ray Computed
18.
Arterioscler Thromb Vasc Biol ; 38(5): 994-1006, 2018 05.
Article in English | MEDLINE | ID: mdl-29496659

ABSTRACT

OBJECTIVE: Macrophages play a central role in various stages of atherosclerotic plaque formation and progression. The local macrophages reportedly proliferate during atherosclerosis, but the pathophysiological significance of macrophage proliferation in this context remains unclear. Here, we investigated the involvement of local macrophage proliferation during atherosclerosis formation and progression using transgenic mice, in which macrophage proliferation was specifically suppressed. APPROACH AND RESULTS: Inhibition of macrophage proliferation was achieved by inducing the expression of cyclin-dependent kinase inhibitor 1B, also known as p27kip, under the regulation of a scavenger receptor promoter/enhancer. The macrophage-specific human p27kip Tg mice were subsequently crossed with apolipoprotein E-deficient mice for the atherosclerotic plaque study. Results showed that a reduced number of local macrophages resulted in marked suppression of atherosclerotic plaque formation and inflammatory response in the plaque. Moreover, fewer local macrophages in macrophage-specific human p27kip Tg mice helped stabilize the plaque, as evidenced by a reduced necrotic core area, increased collagenous extracellular matrix, and thickened fibrous cap. CONCLUSIONS: These results provide direct evidence of the involvement of local macrophage proliferation in formation and progression of atherosclerotic plaques and plaque stability. Thus, control of macrophage proliferation might represent a therapeutic target for treating atherosclerotic diseases.


Subject(s)
Aorta/pathology , Aortitis/prevention & control , Atherosclerosis/prevention & control , Cell Proliferation , Macrophage Activation , Macrophages, Peritoneal/pathology , Plaque, Atherosclerotic , Animals , Aorta/metabolism , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , Fibrosis , Inflammation Mediators/metabolism , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mice, Transgenic , Necrosis , Signal Transduction
19.
Arterioscler Thromb Vasc Biol ; 38(10): 2295-2305, 2018 10.
Article in English | MEDLINE | ID: mdl-30354204

ABSTRACT

Objective- Signaling that activates NFκB (nuclear factor κB) in smooth muscle cells (SMCs) is integral to atherosclerosis and involves reversible ubiquitination that activates proteins downstream of proatherogenic receptors. Deubiquitination of these proteins is mediated by USP20 (ubiquitin-specific protease 20), among other deubiquitinases. We sought to determine whether USP20 activity in SMCs decreases atherosclerosis. Approach and Results- To address this question, we used male Ldlr-/- mice without (control) or with SMC-specific expression of murine USP20 (SMC-USP20-transgenic) or its dominant-negative (DN; C154S/H643Q) mutant (SMC-DN-USP20-transgenic). Before the appearance of intimal macrophages, NFκB activation in aortic medial SMCs was greater in SMC-DN-USP20-transgenic than in control mice. After 16 weeks on a Western diet, SMC-DN-USP20-transgenic mice had 46% greater brachiocephalic artery atheroma area than control mice. Congruently, aortic atherosclerosis assessed en face was 21% greater than control in SMC-DN-USP20-transgenic mice and 13% less than control in SMC-USP20-transgenic mice. In response to TNF (tumor necrosis factor), SMCs from SMC-DN-USP20-transgenic mice showed ≈3-fold greater NFκB activation than control SMCs. Silencing USP20 in SMCs with siRNA (small interfering RNA) augmented NFκB activation by ≈50% in response to either TNF or IL-1ß (interleukin-1ß). Coimmunoprecipitation experiments revealed that USP20 associates with several components of the TNFR1 (TNF receptor-1) signaling pathway, including RIPK1 (receptor-interacting protein kinase 1), a critical checkpoint in TNF-induced NFκB activation and inflammation. TNF evoked ≈2-fold more RIPK1 ubiquitination in SMC-DN-USP20-transgenic than in control SMCs, and RIPK1 was deubiquitinated by purified USP20 in vitro. Conclusions- USP20 attenuates TNF- and IL-1ß-evoked atherogenic signaling in SMCs, by deubiquitinating RIPK1, among other signaling intermediates.


Subject(s)
Aortitis/prevention & control , Atherosclerosis/prevention & control , Endopeptidases/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortitis/enzymology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Endopeptidases/genetics , Female , Hyperplasia , Interleukin-1beta/pharmacology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Neointima , Plaque, Atherosclerotic , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, LDL , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Ubiquitin Thiolesterase , Ubiquitination
20.
Infection ; 47(6): 1059-1063, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31321641

ABSTRACT

Non-typhoidal Salmonella (NTS) spp. causes about 40% of all infective aortitis and it is characterized by high morbidity and mortality. Human infection occurs by fecal-oral transmission through ingestion of contaminated food, milk, or water (inter-human or zoonotic transmission). Approximately 5% of patients with NTS gastroenteritis develop bacteremia and the incidence of extra-intestinal focal infection in NTS bacteremia is about 40%. The organism can reach an extra-intestinal focus through blood dissemination, direct extension from the surrounding organs and direct bacterial inoculation (e.g. invasive medical procedures). Medical and surgical interventions are both needed to successfully control the infection. Here, we report a case of abdominal sub-renal aortitis caused by Salmonella enterica serovar Enteritidis in an 80-year-old man.


Subject(s)
Aorta, Abdominal/surgery , Aortitis/diagnosis , Salmonella Infections/diagnosis , Salmonella enteritidis/isolation & purification , Aged, 80 and over , Aorta, Abdominal/pathology , Aortitis/microbiology , Aortitis/pathology , Aortitis/surgery , Humans , Italy , Male , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella Infections/surgery , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL