Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 347
Filter
1.
Microvasc Res ; 140: 104280, 2022 03.
Article in English | MEDLINE | ID: mdl-34856183

ABSTRACT

Hexarelin, a synthetic growth hormone-releasing peptide, is shown to be protective in cardiovascular diseases such as myocardial infraction and atherosclerosis. However, the functional role of hexarelin in abdominal aortic aneurysm (AAA) remains undefined. The present study determined the effect of hexarelin administration (200 µg/kg twice per day) in a mouse model of elastase-induced abdominal aortic aneurysm. Echocardiography and in situ pictures showed hexarelin decreased infrarenal aorta diameter. Histology staining showed elastin degradation was improved in hexarelin-treated group. Hexarelin rescued smooth muscle cell contractile phenotype with increased α-SMA and decreased MMP2. Furthermore, hexarelin inhibited inflammatory cell infiltration, NLRP3 inflammasome activation and IL-18 production. Particularly, hexarelin suppressed NF-κB signaling pathway which is a key initiator of inflammatory response. These results demonstrated that hexarelin attenuated AAA development by inhibiting SMC phenotype switch and NF-κB signaling mediated inflammatory response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aortic Aneurysm, Abdominal/prevention & control , Cell Plasticity/drug effects , Inflammasomes/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Oligopeptides/pharmacology , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/immunology , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Cytokines/metabolism , Disease Models, Animal , Inflammasomes/metabolism , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phenotype , Signal Transduction , Vascular Remodeling/drug effects
2.
Eur J Pharmacol ; 910: 174490, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34492283

ABSTRACT

Stent-induced vascular injury is manifested by removal of the endothelium and phenotypic changes in the underlying medial smooth muscle cells layer. This results in pathological vascular remodelling primarily contributed to smooth muscle cell proliferation and leads to vessel re-narrowing; neointimal hyperplasia. Current drug-eluting stents release non-selective anti-proliferative drugs such as paclitaxel from the stent surface that not only inhibit growth of smooth muscle cells but also delay endothelial healing, potentially leading to stent thrombosis. This highlights the need for novel bioactive stent coating candidates with the ability to target key events in the pathogenesis of in-stent restenosis. Citric acid, a molecule with anti-coagulant properties, was investigated against L-ascorbic acid, an antioxidant molecule reported to preferentially promote endothelial growth, and paclitaxel, a typically used anti-proliferative stent coating. Citric acid was found to exhibit growth supporting properties on endothelial cells across a range of concentrations that were significantly better than the model stent coating drug paclitaxel and better than the ascorbic acid which inhibited endothelial proliferation at concentrations ≥100 µg/ml. It was demonstrated that a citric acid-paclitaxel combination treatment significantly improves cell viability in comparison to paclitaxel only treated cells, with endothelial cells exhibiting greater cell recovery over smooth muscle cells. Furthermore, cell treatment with citric acid was found to reduce inflammation in a lipopolysaccharide (LPS)-induced in vitro inflammation model by significantly reducing interleukin 6 expression. Thus, this study demonstrates that citric acid is a promising candidate for use as a coating in stents and other endovascular devices.


Subject(s)
Citric Acid/administration & dosage , Drug-Eluting Stents/adverse effects , Thrombosis/prevention & control , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Thrombosis/chemically induced , Thrombosis/immunology , Thrombosis/pathology , Vascular Remodeling/drug effects
3.
Cardiovasc Res ; 117(13): 2624-2638, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34343276

ABSTRACT

Cardiac allograft vasculopathy (CAV) is a pathologic immune-mediated remodelling of the vasculature in transplanted hearts and, by impairing perfusion, is the major cause of late graft loss. Although best understood following cardiac transplantation, similar forms of allograft vasculopathy occur in other vascularized organ grafts and some features of CAV may be shared with other immune-mediated vasculopathies. Here, we describe the incidence and diagnosis, the nature of the vascular remodelling, immune and non-immune contributions to pathogenesis, current therapies, and future areas of research in CAV.


Subject(s)
Coronary Artery Disease/immunology , Coronary Vessels/immunology , Graft Rejection/immunology , Heart Transplantation/adverse effects , Adaptive Immunity , Animals , Coronary Artery Disease/epidemiology , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Graft Rejection/epidemiology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival , Humans , Immunity, Innate , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Risk Factors , Signal Transduction , Treatment Outcome , Vascular Remodeling
4.
Immunobiology ; 226(5): 152124, 2021 09.
Article in English | MEDLINE | ID: mdl-34333403

ABSTRACT

Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling as well as hyper-responsiveness. Thymic stromal lymphopoietin (TSLP), which is a crucial inflammatory cytokine in immune homeostasis, consists of two isoforms, the long isoform lfTSLP and short isoform sfTSLP. The lfTSLP promotes inflammation and plays a pivotal role in asthma pathogenesis, while sfTSLP had been reported to have anti-asthma effects. Experiments have shown that lfTSLP could induce autophagy in hepatocytes. It is unknown whether lfTSLP or sfTSLP could influence autophagy and affect the progression of asthma. Using house dust mite (HDM)-stimulated airway smooth muscle cells as an in vitro model and HDM-induced asthma mice as in vivo model, we found that lfTSLP could induce autophagy and remodeling, while sfTSLP has the reverse effect. Strikingly, sfTSLP treatment in vivo reversed HDM-mediated activation of inflammation and airway remodeling, partly determined by autophagy change. These findings may help us understand the function of TSLP isoforms in the pathogenesis of asthma, and they support the use of drugs targeting sfTSLP and TSLP for asthma treatment.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Cytokines/immunology , Allergens/immunology , Animals , Asthma/blood , Asthma/pathology , Autophagy , Bronchoalveolar Lavage Fluid/cytology , Cell Line , Cytokines/blood , Female , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Lung/pathology , Mice, Inbred C57BL , Myocytes, Smooth Muscle/immunology , Protein Isoforms/immunology , Pyroglyphidae/immunology
5.
Am J Physiol Lung Cell Mol Physiol ; 321(3): L533-L544, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34231388

ABSTRACT

Store-operated calcium entry (SOCE) is involved in the pathogenesis of airway inflammation and remodeling in asthma. Store-operated calcium entry-associated regulatory factor (SARAF) can downregulate SOCE. We sought to investigate the role of SARAF in the regulation of airway inflammation and remodeling in asthma mice models, as well as in the functional regulation of human airway smooth muscle cells (hASMCs). Balb/c mice were sensitized and challenged with ovalbumin to establish the asthma mice models. Mice were transfected with lentivirus, which expressed the SARAF gene + GFP (green fluorescence protein) or the negative control gene + GFP. Airway resistance was measured with the animal pulmonary function system. Airway inflammation and remodeling were evaluated via histological staining. In vitro cultured hASMCs were transfected with scrambled small interfering RNA (siRNA) or SARAF-specific siRNA, respectively. The proliferation, migration rate, hypertrophy, and SOCE activity of hASMCs were examined with Cell Counting Kit-8, wound healing test, bright field imaging, and Ca2+ fluorescence imaging, respectively. SARAF expression was measured by quantitative real-time PCR. Asthma mice models showed decreased SARAF mRNA expression in the lungs. SARAF overexpression attenuated airway inflammation, resistance, and also remodeling. Downregulation of SARAF expression with siRNA promoted the proliferation, migration, hypertrophy, and SOCE activity in hASMCs. SARAF plays a protective role against airway inflammation and remodeling in asthma mice models by blunting SOCE; SARAF may also be a functional regulating factor of hASMCs.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Calcium-Binding Proteins/immunology , Gene Expression Regulation/immunology , Lung/immunology , Membrane Proteins/immunology , Myocytes, Smooth Muscle/immunology , Airway Remodeling/drug effects , Airway Remodeling/genetics , Airway Resistance/drug effects , Airway Resistance/genetics , Airway Resistance/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Calcium-Binding Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Lung/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myocytes, Smooth Muscle/pathology
6.
Cells ; 10(6)2021 06 04.
Article in English | MEDLINE | ID: mdl-34199925

ABSTRACT

Enhanced contractility and migration of airway smooth muscle cells (ASMC) and pulmonary fibroblasts (PF) are part of airway remodeling in asthma. Eosinophils are the central inflammatory cells that participate in airway inflammation. However, the role of asthmatic eosinophils in ASMC and PF contractility, migration, and differentiation to contractile phenotype has not yet been precisely described. A total of 38 individuals were included in this study: 13 steroid-free non-severe allergic asthma (AA) patients, 11 severe non-allergic eosinophilic asthma (SNEA) patients, and 14 healthy subjects (HS). For AA patients and HS groups, a bronchial allergen challenge with D. pteronyssinus was performed. Individual combined cell cultures were prepared from isolated peripheral blood eosinophils and immortalized ASMC or commercial PF cell lines separately. The migration of ASMC and PF was evaluated using wound healing assay and contractility using collagen gel assay. Gene expression of contractile apparatus proteins, COL1A1, COL5A1, and FN, in ASMC and PF was evaluated using qRT-PCR. We found that contractility and migration of ASMC and PF significantly increased after incubation with asthmatic eosinophils compared to HS eosinophils, p < 0.05, and SNEA eosinophils demonstrated the highest effect on contractility of ASMC and migration of both cell lines, p < 0.05. AA and SNEA eosinophils significantly increased gene expression of contractile apparatus proteins, COL1A1 and FN, in both cell lines, p < 0.05. Furthermore, the allergen-activated AA eosinophils significantly increased the contractility of ASMC, and migration and gene expression in ASMC and PF, p < 0.05. Thus, asthmatic eosinophils change ASMC and PF behavior by increasing their contractility and migration, contributing to airway remodeling.


Subject(s)
Asthma/immunology , Cell Movement/immunology , Eosinophils/immunology , Fibroblasts/immunology , Lung/immunology , Muscle Contraction/immunology , Myocytes, Smooth Muscle/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Allergens/immunology , Allergens/pharmacology , Animals , Asthma/pathology , Asthma/physiopathology , Cell Movement/drug effects , Dermatophagoides pteronyssinus/immunology , Eosinophils/pathology , Female , Fibroblasts/pathology , Humans , Lung/pathology , Lung/physiopathology , Male , Middle Aged , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/pathology
7.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34066088

ABSTRACT

Familial hypercholesterolemia (FH) is increasingly associated with inflammation, a phenotype that persists despite treatment with lipid lowering therapies. The alternative C3 complement system (C3), as a key inflammatory mediator, seems to be involved in the atherosclerotic process; however, the relationship between C3 and lipids during plaque progression remains unknown. The aim of the study was to investigate by a systems biology approach the role of C3 in relation to lipoprotein levels during atherosclerosis (AT) progression and to gain a better understanding on the effects of C3 products on the phenotype and function of human lipid-loaded vascular smooth muscle cells (VSMCs). By mass spectrometry and differential proteomics, we found the extracellular matrix (ECM) of human aortas to be enriched in active components of the C3 complement system, with a significantly different proteomic signature in AT segments. Thus, C3 products were more abundant in AT-ECM than in macroscopically normal segments. Furthermore, circulating C3 levels were significantly elevated in FH patients with subclinical coronary AT, evidenced by computed tomographic angiography. However, no correlation was identified between circulating C3 levels and the increase in plaque burden, indicating a local regulation of the C3 in AT arteries. In cell culture studies of human VSMCs, we evidenced the expression of C3, C3aR (anaphylatoxin receptor) and the integrin αMß2 receptor for C3b/iC3b (RT-PCR and Western blot). C3mRNA was up-regulated in lipid-loaded human VSMCs, and C3 protein significantly increased in cell culture supernatants, indicating that the C3 products in the AT-ECM have a local vessel-wall niche. Interestingly, C3a and iC3b (C3 active fragments) have functional effects on VSMCs, significantly reversing the inhibition of VSMC migration induced by aggregated LDL and stimulating cell spreading, organization of F-actin stress fibers and attachment during the adhesion of lipid-loaded human VSMCs. This study, by using a systems biology approach, identified molecular processes involving the C3 complement system in vascular remodeling and in the progression of advanced human atherosclerotic lesions.


Subject(s)
Atherosclerosis/pathology , Complement C3/metabolism , Hyperlipoproteinemia Type II/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Proteome/metabolism , Adult , Atherosclerosis/immunology , Atherosclerosis/metabolism , Case-Control Studies , Cell Adhesion , Cells, Cultured , Female , Humans , Hyperlipoproteinemia Type II/immunology , Hyperlipoproteinemia Type II/metabolism , Male , Middle Aged , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Proteome/analysis , Vascular Remodeling , Wound Healing , Young Adult
8.
Cells ; 10(6)2021 05 25.
Article in English | MEDLINE | ID: mdl-34070626

ABSTRACT

Nipah virus (NiV) is a highly pathogenic zoonotic virus with a broad species tropism, originating in pteropid bats. Human outbreaks of NiV disease occur almost annually, often with high case-fatality rates. The specific events that lead to pathogenesis are not well defined, but the disease has both respiratory and encephalitic components, with relapsing encephalitis occurring in some cases more than a year after initial infection. Several cell types are targets of NiV, dictated by the expression of the ephrin-B2/3 ligand on the cell's outer membrane, which interact with the NiV surface proteins. Vascular endothelial cells (ECs) are major targets of infection. Cytopathic effects (CPE), characterized by syncytia formation and cell death, and an ensuing vasculitis, are a major feature of the disease. Smooth muscle cells (SMCs) of the tunica media that line small blood vessels are infected in humans and animal models of NiV disease, although pathology or histologic changes associated with antigen-positive SMCs have not been reported. To gain an understanding of the possible contributions that SMCs might have in the development of NiV disease, we investigated the susceptibility and potential cytopathogenic changes of human SMCs to NiV infection in vitro. SMCs were permissive for NiV infection and resulted in high titers and prolonged NiV production, despite a lack of cytopathogenicity, and in the absence of detectable ephrin-B2/3. These results indicate that SMC might be important contributors to disease by producing progeny NiV during an infection, without suffering cytopathogenic consequences.


Subject(s)
Endothelial Cells , Henipavirus Infections , Myocytes, Smooth Muscle , Animals , Chlorocebus aethiops , Disease Susceptibility , Endothelial Cells/immunology , Endothelial Cells/virology , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/virology , Nipah Virus , Vero Cells , Virus Replication
9.
Commun Biol ; 4(1): 611, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021256

ABSTRACT

Accumulation of vascular smooth muscle cells (VSMCs) is a hallmark of multiple vascular pathologies, including following neointimal formation after injury and atherosclerosis. However, human VSMCs in advanced atherosclerotic lesions show reduced cell proliferation, extensive and persistent DNA damage, and features of premature cell senescence. Here, we report that stress-induced premature senescence (SIPS) and stable expression of a telomeric repeat-binding factor 2 protein mutant (TRF2T188A) induce senescence of human VSMCs, associated with persistent telomeric DNA damage. VSMC senescence is associated with formation of micronuclei, activation of cGAS-STING cytoplasmic sensing, and induction of multiple pro-inflammatory cytokines. VSMC-specific TRF2T188A expression in a multicolor clonal VSMC-tracking mouse model shows no change in VSMC clonal patches after injury, but an increase in neointima formation, outward remodeling, senescence and immune/inflammatory cell infiltration or retention. We suggest that persistent telomere damage in VSMCs inducing cell senescence has a major role in driving persistent inflammation in vascular disease.


Subject(s)
Atherosclerosis/pathology , Cellular Senescence , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neointima/pathology , Telomere/pathology , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cell Proliferation , Cells, Cultured , DNA Damage , Disease Models, Animal , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/physiology , Muscle Proteins/physiology , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Neointima/etiology , Neointima/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 2/metabolism
10.
Respir Res ; 22(1): 158, 2021 May 22.
Article in English | MEDLINE | ID: mdl-34022896

ABSTRACT

BACKGROUND: RORγt is a transcription factor that enables elaboration of Th17-associated cytokines (including IL-17 and IL-22) and is proposed as a pharmacological target for severe asthma. METHODS: IL-17 immunohistochemistry was performed in severe asthma bronchial biopsies (specificity confirmed with in situ hybridization). Primary human small airway epithelial cells in air liquid interface and primary bronchial smooth muscle cells were stimulated with recombinant human IL-17 and/or IL-22 and pro-inflammatory cytokines measured. Balb/c mice were challenged intratracheally with IL-17 and/or IL-22 and airway hyperreactivity, pro-inflammatory cytokines and airway neutrophilia measured. Balb/c mice were sensitized intraperitoneally and challenged intratracheally with house dust mite extract and the effect of either a RORγt inhibitor (BIX119) or an anti-IL-11 antibody assessed on airway hyperreactivity, pro-inflammatory cytokines and airway neutrophilia measured. RESULTS: We confirmed in severe asthma bronchial biopsies both the presence of IL-17-positive lymphocytes and that an IL-17 transcriptome profile in a severe asthma patient sub-population. Both IL-17 and IL-22 stimulated the release of pro-inflammatory cytokine and chemokine release from primary human lung cells and in mice. Furthermore, IL-22 in combination with IL-17, but neither alone, elicits airway hyperresponsiveness (AHR) in naïve mice. A RORγt inhibitor specifically blocked both IL-17 and IL-22, AHR and neutrophilia in a mouse house dust mite model unlike other registered or advanced pipeline modes of action. Full efficacy versus these parameters was associated with 90% inhibition of IL-17 and 50% inhibition of IL-22. In contrast, anti-IL-17 also blocked IL-17, but not IL-22, AHR or neutrophilia. Moreover, the deregulated genes in the lungs from these mice correlated well with deregulated genes from severe asthma biopsies suggesting that this model recapitulates significant severe asthma-relevant biology. Furthermore, these genes were reversed upon RORγt inhibition in the HDM model. Cell deconvolution suggested that the responsible cells were corticosteroid insensitive γδ-T-cells. CONCLUSION: These data strongly suggest that both IL-17 and IL-22 are required for Th2-low endotype associated biology and that a RORγt inhibitor may provide improved clinical benefit in a severe asthma sub-population of patients by blocking both IL-17 and IL-22 biology compared with blocking IL-17 alone.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Interleukin-17/metabolism , Interleukins/antagonists & inhibitors , Lung/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Th17 Cells/drug effects , Adolescent , Adult , Aged , Animals , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Humans , Interleukins/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Male , Mice, Inbred BALB C , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pyroglyphidae/immunology , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult , Interleukin-22
11.
Pharmacol Res ; 165: 105447, 2021 03.
Article in English | MEDLINE | ID: mdl-33516832

ABSTRACT

Pyroptosis is a form of programmed cell death activated by various stimuli and is characterized by inflammasome assembly, membrane pore formation, and the secretion of inflammatory cytokines (IL-1ß and IL-18). Atherosclerosis-related risk factors, including oxidized low-density lipoprotein (ox-LDL) and cholesterol crystals, have been shown to promote pyroptosis through several mechanisms that involve ion flux, ROS, endoplasmic reticulum stress, mitochondrial dysfunction, lysosomal rupture, Golgi function, autophagy, noncoding RNAs, post-translational modifications, and the expression of related molecules. Pyroptosis of endothelial cells, macrophages, and smooth muscle cells in the vascular wall can induce plaque instability and accelerate atherosclerosis progression. In this review, we focus on the pathogenesis, influence, and therapy of pyroptosis in atherosclerosis and provide novel ideas for suppressing pyroptosis and the progression of atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Endothelial Cells/immunology , Immunity, Cellular/immunology , Inflammation Mediators/immunology , Pyroptosis/immunology , Animals , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , Macrophages/immunology , Macrophages/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism
12.
Arterioscler Thromb Vasc Biol ; 41(2): 796-807, 2021 02.
Article in English | MEDLINE | ID: mdl-33380173

ABSTRACT

OBJECTIVE: The aim of this study was to unravel mechanisms whereby deficiency of the transcription factor Id3 (inhibitor of differentiation 3) leads to metabolic dysfunction in visceral obesity. We investigated the impact of loss of Id3 on hyaluronic acid (HA) production by the 3 HAS isoenzymes (HA synthases; -1, -2, and -3) and on obesity-induced adipose tissue (AT) accumulation of proinflammatory B cells. Approach and Results: Male Id3-/- mice and respective wild-type littermate controls were fed a 60% high-fat diet for 4 weeks. An increase in inflammatory B2 cells was detected in Id3-/- epididymal AT. HA accumulated in epididymal AT of high-fat diet-fed Id3-/- mice and circulating levels of HA were elevated. Has2 mRNA expression was increased in epididymal AT of Id3-/- mice. Luciferase promoter assays showed that Id3 suppressed Has2 promoter activity, while loss of Id3 stimulated Has2 promoter activity. Functionally, HA strongly promoted B2 cell adhesion in the AT and on cultured vascular smooth muscle cells of Id3-/- mice, an effect sensitive to hyaluronidase. CONCLUSIONS: Our data demonstrate that loss of Id3 increases Has2 expression in the epididymal AT, thereby promoting HA accumulation. In turn, elevated HA content promotes HA-dependent binding of B2 cells and an increase in the B2 cells in the AT, which contributes to AT inflammation.


Subject(s)
Adipose Tissue/metabolism , B-Lymphocytes/metabolism , Hyaluronan Synthases/metabolism , Hyaluronic Acid/biosynthesis , Inhibitor of Differentiation Proteins/metabolism , Panniculitis/metabolism , Adipose Tissue/immunology , Animals , B-Lymphocytes/immunology , Cell Adhesion , Cells, Cultured , Coculture Techniques , Diet, High-Fat , Disease Models, Animal , Hyaluronan Synthases/genetics , Inhibitor of Differentiation Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Panniculitis/genetics , Panniculitis/immunology , Phenotype , Signal Transduction , Up-Regulation
13.
Cardiovasc Res ; 117(3): 930-941, 2021 02 22.
Article in English | MEDLINE | ID: mdl-32243494

ABSTRACT

AIMS: Uromodulin is produced exclusively in the kidney and secreted into both urine and blood. Serum levels of uromodulin are correlated with kidney function and reduced in chronic kidney disease (CKD) patients, but physiological functions of serum uromodulin are still elusive. This study investigated the role of uromodulin in medial vascular calcification, a key factor associated with cardiovascular events and mortality in CKD patients. METHODS AND RESULTS: Experiments were performed in primary human (HAoSMCs) and mouse (MOVAS) aortic smooth muscle cells, cholecalciferol overload and subtotal nephrectomy mouse models and serum from CKD patients. In three independent cohorts of CKD patients, serum uromodulin concentrations were inversely correlated with serum calcification propensity. Uromodulin supplementation reduced phosphate-induced osteo-/chondrogenic transdifferentiation and calcification of HAoSMCs. In human serum, pro-inflammatory cytokines tumour necrosis factor α (TNFα) and interleukin-1ß (IL-1ß) co-immunoprecipitated with uromodulin. Uromodulin inhibited TNFα and IL-1ß-induced osteo-/chondrogenic signalling and activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated ß cells (NF-kB) as well as phosphate-induced NF-kB-dependent transcriptional activity in HAoSMCs. In vivo, adeno-associated virus (AAV)-mediated overexpression of uromodulin ameliorated vascular calcification in mice with cholecalciferol overload. Conversely, cholecalciferol overload-induced vascular calcification was aggravated in uromodulin-deficient mice. In contrast, uromodulin overexpression failed to reduce vascular calcification during renal failure in mice. Carbamylated uromodulin was detected in serum of CKD patients and uromodulin carbamylation inhibited its anti-calcific properties in vitro. CONCLUSIONS: Uromodulin counteracts vascular osteo-/chondrogenic transdifferentiation and calcification, at least in part, through interference with cytokine-dependent pro-calcific signalling. In CKD, reduction and carbamylation of uromodulin may contribute to vascular pathology.


Subject(s)
Cell Transdifferentiation , Cytokines/metabolism , Inflammation Mediators/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Renal Insufficiency, Chronic/blood , Uromodulin/blood , Vascular Calcification/prevention & control , Adult , Aged , Animals , Aorta/immunology , Aorta/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Chondrogenesis , Cytokines/genetics , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Osteogenesis , Phenotype , Protein Carbamylation , Renal Insufficiency, Chronic/immunology , Signal Transduction , Uromodulin/genetics , Uromodulin/pharmacology , Vascular Calcification/blood , Vascular Calcification/immunology , Young Adult
14.
Methods Mol Biol ; 2223: 217-236, 2021.
Article in English | MEDLINE | ID: mdl-33226598

ABSTRACT

Cellular inflammation, with elevated levels of Th1/Th2 cytokines, airway mucus hypersecretion, and thickening of the airway smooth muscle, are characteristic features of the allergic lung. Assessment of pathophysiological changes in allergic lungs serves as an important tool to determine disease progression and understand the underlying mechanisms of pathogenesis. This can be achieved by evaluating the lung tissue for inflammation and airway structural changes along with the measurement of important pro-inflammatory mediators such as Th1/Th2 cytokines and eotaxins. This chapter describes procedures to histologically evaluate inflammatory and pathological changes observed during allergic airway inflammation using lung tissue from mice.


Subject(s)
Allergens/administration & dosage , Asthma/immunology , Lung/immunology , Respiratory Hypersensitivity/immunology , Staining and Labeling/methods , Th1-Th2 Balance , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/biosynthesis , Cytokines/immunology , Disease Progression , Lung/metabolism , Lung/pathology , Mice , Microtomy/methods , Mucus/immunology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Paraffin Embedding/methods , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
15.
Front Immunol ; 11: 599415, 2020.
Article in English | MEDLINE | ID: mdl-33324416

ABSTRACT

The pathobiology of atherosclerotic disease requires further elucidation to discover new approaches to address its high morbidity and mortality. To date, over 17 million cardiovascular-related deaths have been reported annually, despite a multitude of surgical and nonsurgical interventions and advances in medical therapy. Existing strategies to prevent disease progression mainly focus on management of risk factors, such as hypercholesterolemia. Even with optimum current medical therapy, recurrent cardiovascular events are not uncommon in patients with atherosclerosis, and their incidence can reach 10-15% per year. Although treatments targeting inflammation are under investigation and continue to evolve, clinical breakthroughs are possible only if we deepen our understanding of vessel wall pathobiology. Vascular smooth muscle cells (VSMCs) are one of the most abundant cells in vessel walls and have emerged as key players in disease progression. New technologies, including in situ hybridization proximity ligation assays, in vivo cell fate tracing with the CreERT2-loxP system and single-cell sequencing technology with spatial resolution, broaden our understanding of the complex biology of these intriguing cells. Our knowledge of contractile and synthetic VSMC phenotype switching has expanded to include macrophage-like and even osteoblast-like VSMC phenotypes. An increasing body of data suggests that VSMCs have remarkable plasticity and play a key role in cell-to-cell crosstalk with endothelial cells and immune cells during the complex process of inflammation. These are cells that sense, interact with and influence the behavior of other cellular components of the vessel wall. It is now more obvious that VSMC plasticity and the ability to perform nonprofessional phagocytic functions are key phenomena maintaining the inflammatory state and senescent condition and actively interacting with different immune competent cells.


Subject(s)
Atherosclerosis/immunology , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/immunology , Vasculitis/immunology , Animals , Atherosclerosis/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Vasculitis/pathology
16.
Front Immunol ; 11: 1899, 2020.
Article in English | MEDLINE | ID: mdl-32983111

ABSTRACT

Background: Infection/inflammation is an important causal factor in spontaneous preterm birth (sPTB). Most mechanistic studies have concentrated on the role of bacteria, with limited focus on the role of viruses in sPTB. Murine studies support a potential multi-pathogen aetiology in which a double or sequential hit of both viral and bacterial pathogens leads to a higher risk preterm labour. This study aimed to determine the effect of viral priming on bacterial induced inflammation in human in vitro models of ascending and haematogenous infection. Methods: Vaginal epithelial cells, and primary amnion epithelial cells and myocytes were used to represent cell targets of ascending infection while interactions between peripheral blood mononuclear cells (PBMCs) and placental explants were used to model systemic infection. To model the effect of viral priming upon the subsequent response to bacterial stimuli, each cell type was stimulated first with a TLR3 viral agonist, and then with either a TLR2 or TLR2/6 agonist, and responses compared to those of each agonist alone. Immunoblotting was used to detect cellular NF-κB, AP-1, and IRF-3 activation. Cellular TLR3, TLR2, and TLR6 mRNA was quantified by RT-qPCR. Immunoassays were used to measure supernatant cytokine, chemokine and PGE2 concentrations. Results: TLR3 ("viral") priming prior to TLR2/6 agonist ("bacterial") exposure augmented the pro-inflammatory, pro-labour response in VECs, AECs, myocytes and PBMCs when compared to the effects of agonists alone. In contrast, enhanced anti-inflammatory cytokine production (IL-10) was observed in placental explants. Culturing placental explants in conditioned media derived from PBMCs primed with a TLR3 agonist enhanced TLR2/6 agonist stimulated production of IL-6 and IL-8, suggesting a differential response by the placenta to systemic inflammation compared to direct infection as a result of haematogenous spread. TLR3 agonism generally caused increased mRNA expression of TLR3 and TLR2 but not TLR6. Conclusion: This study provides human in vitro evidence that viral infection may increase the susceptibility of women to bacterial-induced sPTB. Improved understanding of interactions between viral and bacterial components of the maternal microbiome and host immune response may offer new therapeutic options, such as antivirals for the prevention of PTB.


Subject(s)
Amnion/drug effects , Immunologic Factors/pharmacology , Myometrium/drug effects , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/virology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 3/agonists , Toll-Like Receptor 6/agonists , Vagina/drug effects , Amnion/immunology , Amnion/metabolism , Cell Line , Cytokines/metabolism , Dinoprostone/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Host-Pathogen Interactions , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Myometrium/immunology , Myometrium/metabolism , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/metabolism , Signal Transduction , Tissue Culture Techniques , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 6/genetics , Toll-Like Receptor 6/metabolism , Vagina/immunology , Vagina/metabolism
17.
Front Immunol ; 11: 1798, 2020.
Article in English | MEDLINE | ID: mdl-32903501

ABSTRACT

Interleukin (IL)-33 plays important roles in pulmonary immune responses and lung diseases including asthma and chronic obstructive pulmonary disease (COPD). There is substantial interest in identifying and characterizing cellular sources vs. targets of IL-33, and downstream signaling pathways involved in disease pathophysiology. While epithelial and immune cells have largely been the focus, in this review, we summarize current knowledge of expression, induction, and function of IL-33 and its receptor ST2 in non-hematopoietic lung cells in the context of health and disease. Under basal conditions, epithelial cells and endothelial cells are thought to be the primary resident cell types that express high levels of IL-33 and serve as ligand sources compared to mesenchymal cells (smooth muscle cells and fibroblasts). Under inflammatory conditions, IL-33 expression is increased in most non-hematopoietic lung cells, including epithelial, endothelial, and mesenchymal cells. In comparison to its ligand, the receptor ST2 shows low expression levels at baseline but similar to IL-33, ST2 expression is upregulated by inflammation in these non-hematopoietic lung cells which may then participate in chronic inflammation both as sources and autocrine/paracrine targets of IL-33. Downstream effects of IL-33 may occur via direct receptor activation or indirect interactions with the immune system, overall contributing to lung inflammation, airway hyper-responsiveness and remodeling (proliferation and fibrosis). Accordingly from a therapeutic perspective, targeting IL-33 and/or its receptor in non-hematopoietic lung cells becomes relevant.


Subject(s)
Interleukin-33/immunology , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Animals , Endothelial Cells/immunology , Epithelial Cells/immunology , Fibroblasts/immunology , Humans , Myocytes, Smooth Muscle/immunology
18.
Curr Opin Pharmacol ; 51: 50-58, 2020 04.
Article in English | MEDLINE | ID: mdl-32836013

ABSTRACT

Asthma is an obstructive inflammatory airway disease. Airway obstruction is mediated by hyperresponsive airway smooth muscle cell contraction, which is induced and compounded by inflammation caused by T lymphocytes. One important signal transduction pathway that is involved in the activation of these cell types involves the generation of a lipid second messenger known as diacylglycerol (DAG). DAG levels are controlled in cells by a negative regulator known as DAG kinase (DGK). In this review, we discuss how the DAG signaling pathway attenuates the pathological function of immune cells and airway smooth muscle cells in allergic airway disease and asthma. Furthermore, we discuss how the enhancement of the DAG signaling pathway through the inhibition of DGK may represent a novel therapeutic strategy for these diseases.


Subject(s)
Diacylglycerol Kinase/metabolism , Hypersensitivity/enzymology , Lung Diseases/enzymology , Myocytes, Smooth Muscle/enzymology , T-Lymphocytes/enzymology , Animals , Anti-Asthmatic Agents/administration & dosage , Diacylglycerol Kinase/immunology , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Lung Diseases/drug therapy , Lung Diseases/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
19.
Curr Opin Pharmacol ; 51: 11-18, 2020 04.
Article in English | MEDLINE | ID: mdl-32361678

ABSTRACT

Chronic pulmonary diseases, including chronic obstructive pulmonary disease (COPD) and asthma, are major causes of death and reduced quality of life. Characteristic of chronic pulmonary disease is excessive lung inflammation that occurs in response to exposure to inhaled irritants, chemicals, and allergens. Chronic inflammation leads to remodeling of the airways that includes excess mucus secretion, proliferation of smooth muscle cells, increased deposition of extracellular matrix proteins and fibrosis. Protein kinases have been implicated in mediating inflammatory signals and airway remodeling associated with reduced lung function in chronic pulmonary disease. This review will highlight the role of protein kinases in the lung during chronic inflammation and examine opportunities to use protein kinase inhibitors for the treatment of chronic pulmonary diseases.


Subject(s)
Lung Diseases, Obstructive/drug therapy , Lung Diseases, Obstructive/enzymology , Lung/drug effects , Lung/enzymology , Protein Kinase Inhibitors/therapeutic use , Airway Remodeling/drug effects , Airway Remodeling/physiology , Animals , Humans , Lung/immunology , Lung Diseases, Obstructive/immunology , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Muscle, Smooth/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , Protein Kinase Inhibitors/pharmacology , Treatment Outcome
20.
Int J Mol Sci ; 21(5)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155894

ABSTRACT

Eosinophils infiltration and releasing TGF-ß1 in the airways has been implicated in the pathogenesis of asthma, especially during acute episodes provoked by an allergen. TGF-ß1 is a major mediator involved in pro-inflammatory responses and fibrotic tissue remodeling in asthma. We aimed to evaluate the effect of in vivo allergen-activated eosinophils on the expression of COL1A1 and FN in ASM cells in asthma. A total of 12 allergic asthma patients and 11 healthy subjects were examined. All study subjects underwent bronchial challenge with D. pteronyssinus allergen. Eosinophils from peripheral blood were isolated before and 24 h after the bronchial allergen challenge using high-density centrifugation and magnetic separation. Individual co-cultures of blood eosinophils and immortalized human ASM cells were prepared. The TGF-ß1 concentration in culture supernatants was analyzed using ELISA. Gene expression was analyzed using qRT-PCR. Eosinophils integrins were suppressed with linear RGDS peptide before co-culture with ASM cells. Results: The expression of TGF-ß1 in asthmatic eosinophils significantly increased over non-activated asthmatic eosinophils after allergen challenge, p < 0.001. The TGF-ß1 concentration in culture supernatants was significantly higher in samples with allergen-activated asthmatic eosinophils compared to baseline, p < 0.05. The effect of allergen-activated asthmatic eosinophils on the expression of TGF-ß1, COL1A1, and FN in ASM cells was more significant compared to non-activated eosinophils, p < 0.05, however, no difference was found on WNT-5A expression. The incubation of allergen-activated asthmatic eosinophils with RGDS peptide was more effective compared to non-activated eosinophils as the gene expression in ASM cells was downregulated equally to the same level as healthy eosinophils.


Subject(s)
Asthma/pathology , Bronchial Provocation Tests/adverse effects , Collagen Type I/metabolism , Eosinophils/immunology , Fibronectins/metabolism , Myocytes, Smooth Muscle/immunology , Respiratory System/immunology , Transforming Growth Factor beta1/metabolism , Adult , Asthma/chemically induced , Asthma/immunology , Asthma/metabolism , Case-Control Studies , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Eosinophils/drug effects , Female , Fibronectins/genetics , Gene Expression Regulation , Humans , Male , Myocytes, Smooth Muscle/drug effects , Respiratory System/drug effects , Signal Transduction , Transforming Growth Factor beta1/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...