Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Circ Res ; 125(2): 223-241, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31079549

ABSTRACT

RATIONALE: Transplantation-accelerated arteriosclerosis is one of the major challenges for long-term survival of patients with solid organ transplantation. Although stem/progenitor cells have been implicated to participate in this process, the cells of origin and underlying mechanisms have not been fully defined. OBJECTIVE: The objective of our study was to investigate the role of c-Kit lineage cells in allograft-induced neointima formation and to explore the mechanisms underlying this process. METHODS AND RESULTS: Using an inducible lineage tracing Kit-CreER;Rosa26-tdTomato mouse model, we observed that c-Kit is expressed in multiple cell types in the blood vessels, rather than a specific stem/progenitor cell marker. We performed allograft transplantation between different donor and recipient mice, as well as bone marrow transplantation experiments, demonstrating that recipient c-Kit+ cells repopulate neointimal smooth muscle cells (SMCs) and leukocytes, and contribute to neointima formation in an allograft transplantation model. c-Kit-derived SMCs originate from nonbone marrow tissues, whereas bone marrow-derived c-Kit+ cells mainly generate CD45+ leukocytes. However, the exact identity of c-Kit lineage cells contributing to neointimal SMCs remains unclear. ACK2 (anti-c-Kit antibody), which specifically binds and blocks c-Kit function, ameliorates allograft-induced arteriosclerosis. Stem cell factor and TGF (transforming growth factor)-ß1 levels were significantly increased in blood and neointimal lesions after allograft transplantation, by which stem cell factor facilitated c-Kit+ cell migration through the stem cell factor/c-Kit axis and downstream activation of small GTPases, MEK (mitogen-activated protein kinase kinase)/ERK (extracellular signal-regulated kinase)/MLC (myosin light chain), and JNK (c-Jun N-terminal kinase)/c-Jun signaling pathways, whereas TGF-ß1 induces c-Kit+ cell differentiation into SMCs via HK (hexokinase)-1-dependent metabolic reprogramming and a possible downstream O-GlcNAcylation of myocardin and serum response factor. CONCLUSIONS: Our findings provide evidence that recipient c-Kit lineage cells contribute to vascular remodeling in an allograft transplantation model, in which the stem cell factor/c-Kit axis is responsible for cell migration and HK-1-dependent metabolic reprogramming for SMC differentiation.


Subject(s)
Arteriosclerosis/therapy , Cell Movement , Myocytes, Smooth Muscle/physiology , Animals , Aorta/physiology , Aorta/transplantation , Cells, Cultured , Cellular Reprogramming , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Regeneration , Stem Cell Factor/metabolism , Tunica Intima/cytology , Tunica Intima/physiology
2.
Arterioscler Thromb Vasc Biol ; 38(2): 425-437, 2018 02.
Article in English | MEDLINE | ID: mdl-29284609

ABSTRACT

OBJECTIVE: DKK3 (dickkopf 3), a 36-kD secreted glycoprotein, has been shown to be involved in the differentiation of partially reprogrammed cells and embryonic stem cells to smooth muscle cells (SMCs), but little is known about its involvement in vascular disease. This study aims to assess the effects of DKK3 on atherosclerotic plaque composition. APPROACH AND RESULTS: In the present study, we used a murine model of atherosclerosis (ApoE-/-) in conjunction with DKK3-/- and performed tandem stenosis of the carotid artery to evaluate atherosclerotic plaque development. We found that the absence of DKK3 leads to vulnerable atherosclerotic plaques, because of a reduced number of SMCs and reduced matrix protein deposition, as well as increased hemorrhage and macrophage infiltration. Further in vitro studies revealed that DKK3 can induce differentiation of Sca1+ (stem cells antigen 1) vascular progenitors and fibroblasts into SMCs via activation of the TGF-ß (transforming growth factor-ß)/ATF6 (activating transcription factor 6) and Wnt signaling pathways. Finally, we assessed the therapeutic potential of DKK3 in mouse and rabbit models and found that DKK3 altered the atherosclerotic plaque content via increasing SMC numbers and reducing vascular inflammation. CONCLUSIONS: Cumulatively, we provide the first evidence that DKK3 is a potent SMC differentiation factor, which might have a therapeutic effect in reducing intraplaque hemorrhage related to atherosclerotic plaque phenotype.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Carotid Stenosis/metabolism , Cell Transdifferentiation , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Stem Cells/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Adaptor Proteins, Signal Transducing , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Ataxin-1/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Stenosis/genetics , Carotid Stenosis/pathology , Cells, Cultured , Chemokines , Disease Models, Animal , Female , Fibroblasts/pathology , Hemorrhage/genetics , Hemorrhage/metabolism , Hemorrhage/pathology , Hemorrhage/prevention & control , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Rabbits , Stem Cells/pathology , Transforming Growth Factor beta1/metabolism , Wnt Signaling Pathway
3.
Arterioscler Thromb Vasc Biol ; 37(11): 2114-2127, 2017 11.
Article in English | MEDLINE | ID: mdl-28935755

ABSTRACT

OBJECTIVE: Leptin is an adipokine initially thought to be a metabolic factor. Recent publications have shown its roles in inflammation and vascular disease, to which Sca-1+ vascular progenitor cells within the vessel wall may contribute. We sought to elucidate the effects of leptin on Sca-1+ progenitor cells migration and neointimal formation and to understand the underlying mechanisms. APPROACH AND RESULTS: Sca-1+ progenitor cells from the vessel wall of Lepr+/+ and Lepr-/- mice were cultured and purified. The migration of Lepr+/+ Sca-1+ progenitor cells in vitro was markedly induced by leptin. Western blotting and kinase assays revealed that leptin induced the activation of phosphorylated signal transducer and activator of transcription 3, phosphorylated extracellular signal-regulated kinases 1/2, pFAK (phosphorylated focal adhesion kinase), and Rac1 (ras-related C3 botulinum toxin substrate 1)/Cdc42 (cell division control protein 42 homolog). In a mouse femoral artery guidewire injury model, an increased expression of leptin in both injured vessels and serum was observed 24 hours post-surgery. RFP (red fluorescent protein)-Sca-1+ progenitor cells in Matrigel were applied to the adventitia of the injured femoral artery. RFP+ cells were observed in the intima 24 hours post-surgery, subsequently increasing neointimal lesions at 2 weeks when compared with the arteries without seeded cells. This increase was reduced by pre-treatment of Sca-1+ cells with a leptin antagonist. Guidewire injury could only induce minor neointima in Lepr-/- mice 2 weeks post-surgery. However, transplantation of Lepr+/+ Sca-1+ progenitor cells into the adventitial side of injured artery in Lepr-/- mice significantly enhanced neointimal formation. CONCLUSIONS: Upregulation of leptin levels in both the vessel wall and the circulation after vessel injury promoted the migration of Sca-1+ progenitor cells via leptin receptor-dependent signal transducer and activator of transcription 3- Rac1/Cdc42-ERK (extracellular signal-regulated kinase)-FAK pathways, which enhanced neointimal formation.


Subject(s)
Antigens, Ly/metabolism , Cell Movement , Leptin/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Stem Cells/metabolism , Vascular System Injuries/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Focal Adhesion Kinase 1/metabolism , Genetic Predisposition to Disease , Male , Mice, Knockout , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/transplantation , Neuropeptides/metabolism , Phenotype , Phosphorylation , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Stem Cell Transplantation , Stem Cells/pathology , Time Factors , Up-Regulation , Vascular System Injuries/genetics , Vascular System Injuries/pathology , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
4.
Stem Cell Reports ; 9(2): 681-696, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28757161

ABSTRACT

Adventitial progenitor cells, including SCA-1+ and mesenchymal stem cells, are believed to be important in vascular remodeling. It has been shown that SCA-1+ progenitor cells are involved in neointimal hyperplasia of vein grafts, but little is known concerning their involvement in hyperlipidemia-induced atherosclerosis. We employed single-cell sequencing technology on primary adventitial mouse SCA-1+ cells from wild-type and atherosclerotic-prone (ApoE-deficient) mice and found that a group of genes controlling cell migration and matrix protein degradation was highly altered. Adventitial progenitors from ApoE-deficient mice displayed an augmented migratory potential both in vitro and in vivo. This increased migratory ability was mimicked by lipid loading to SCA-1+ cells. Furthermore, we show that lipid loading increased miRNA-29b expression and induced sirtuin-1 and matrix metalloproteinase-9 levels to promote cell migration. These results provide direct evidence that blood cholesterol levels influence vascular progenitor cell function, which could be a potential target cell for treatment of vascular disease.


Subject(s)
Ataxin-1/genetics , Cell Movement/genetics , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Stem Cells/metabolism , Animals , Apolipoproteins E/deficiency , Ataxin-1/metabolism , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers , Cell Differentiation/genetics , Cholesterol, LDL/metabolism , Computational Biology/methods , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Hyperlipidemias/blood , Immunohistochemistry , Inflammation Mediators/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Stem Cells/cytology
5.
Stem Cells ; 34(9): 2368-80, 2016 09.
Article in English | MEDLINE | ID: mdl-27300479

ABSTRACT

Recent studies have shown that Sca-1(+) (stem cell antigen-1) stem/progenitor cells within blood vessel walls may contribute to neointima formation, but the mechanism behind their recruitment has not been explored. In this work Sca-1(+) progenitor cells were cultivated from mouse vein graft tissue and found to exhibit increased migration when cocultured with smooth muscle cells (SMCs) or when treated with SMC-derived conditioned medium. This migration was associated with elevated levels of chemokines, CCL2 (chemokine (C-C motif) ligand 2) and CXCL1 (chemokine (C-X-C motif) ligand 1), and their corresponding receptors on Sca-1(+) progenitors, CCR2 (chemokine (C-C motif) receptor 2) and CXCR2 (chemokine (C-X-C motif) receptor 2), which were also upregulated following SMC conditioned medium treatment. Knockdown of either receptor in Sca-1(+) progenitors significantly inhibited cell migration. The GTPases Cdc42 and Rac1 were activated by both CCL2 and CXCL1 stimulation and p38 phosphorylation was increased. However, only Rac1 inhibition significantly reduced migration and p38 phosphorylation. After Sca-1(+) progenitors labeled with green fluorescent protein (GFP) were applied to the adventitial side of wire-injured mouse femoral arteries, a large proportion of GFP-Sca-1(+) -cells were observed in neointimal lesions, and a marked increase in neointimal lesion formation was seen 1 week post-operation. Interestingly, Sca-1(+) progenitor migration from the adventitia to the neointima was abrogated and neointima formation diminished in a wire injury model using CCL2(-/-) mice. These findings suggest vascular stem/progenitor cell migration from the adventitia to the neointima can be induced by SMC release of chemokines which act via CCR2/Rac1/p38 and CXCR2/Rac1/p38 signaling pathways. Stem Cells 2016;34:2368-2380.


Subject(s)
Cell Movement , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/pathology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Antigens, Ly/metabolism , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Receptors, CCR2 , Receptors, Interleukin-8B/metabolism , Signal Transduction/drug effects , cdc42 GTP-Binding Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
6.
Arterioscler Thromb Vasc Biol ; 34(10): 2184-90, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25012135

ABSTRACT

Mechanical forces have long been known to play a role in the maintenance of vascular homeostasis in the mature animal and in developmental regulation in the fetus. More recently, it has been shown that stem cells play a role in vascular repair and remodeling in response to biomechanical stress. Laminar shear stress can directly activate growth factor receptors on stem/progenitor cells, initiating signaling pathways leading toward endothelial cell differentiation. Cyclic strain can stimulate stem cell differentiation toward smooth muscle lineages through different mechanisms. In vivo, blood flow in the coronary artery is significantly altered after stenting, leading to changes in biomechanical forces on the vessel wall. This disruption may activate stem cell differentiation into a variety of cells and cause delayed re-endothelialization. Based on progress in the research field, the present review aims to explore the role of mechanical forces in stem cell differentiation both in vivo and in vitro and to examine what this means for the application of stem cells in the clinic, in tissue engineering, and for the management of aberrant stem cell contribution to disease.


Subject(s)
Atherosclerosis/pathology , Blood Vessels/pathology , Cell Differentiation , Cell Lineage , Mechanotransduction, Cellular , Stem Cells/pathology , Animals , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Atherosclerosis/therapy , Blood Vessels/metabolism , Blood Vessels/physiopathology , Cell Proliferation , Humans , Phenotype , Regeneration , Regional Blood Flow , Stem Cells/metabolism , Stress, Mechanical
7.
Proc Natl Acad Sci U S A ; 109(43): 17597-602, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23045674

ABSTRACT

Prostacyclin is an antithrombotic hormone produced by the endothelium, whose production is dependent on cyclooxygenase (COX) enzymes of which two isoforms exist. It is widely believed that COX-2 drives prostacyclin production and that this explains the cardiovascular toxicity associated with COX-2 inhibition, yet the evidence for this relies on indirect evidence from urinary metabolites. Here we have used a range of experimental approaches to explore which isoform drives the production of prostacyclin in vitro and in vivo. Our data show unequivocally that under physiological conditions it is COX-1 and not COX-2 that drives prostacyclin production in the cardiovascular system, and that urinary metabolites do not reflect prostacyclin production in the systemic circulation. With the idea that COX-2 in endothelium drives prostacyclin production in healthy individuals removed, we must seek new answers to why COX-2 inhibitors increase the risk of cardiovascular events to move forward with drug discovery and to enable more informed prescribing advice.


Subject(s)
Cardiovascular System/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Epoprostenol/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
PLoS One ; 7(2): e31228, 2012.
Article in English | MEDLINE | ID: mdl-22359578

ABSTRACT

OBJECTIVE: To measure the elongation and compliance of endothelial cells subjected to different patterns of shear stress in vitro, and to compare these parameters with the elongation and compliance of endothelial cells from different regions of the intact aorta. MATERIALS AND METHODS: Porcine aortic endothelial cells were cultured for 6 days under static conditions or on an orbital shaker. The shaker generated a wave of medium, inducing pulsatile shear stress with a preferred orientation at the edge of the well or steadier shear stress with changing orientation at its centre. The topography and compliance of these cells and cells from the inner and outer curvature of ex vivo porcine aortic arches were measured by scanning ion conductance microscopy (SICM). RESULTS: Cells cultured under oriented shear stress were more elongated and less compliant than cells grown under static conditions or under shear stress with no preferred orientation. Cells from the outer curvature of the aorta were more elongated and less compliant than cells from the inner curvature. CONCLUSION: The elongation and compliance of cultured endothelial cells vary according to the pattern of applied shear stress, and are inversely correlated. A similar inverse correlation occurs in the aortic arch, with variation between regions thought to experience different haemodynamic stresses.


Subject(s)
Aorta/cytology , Endothelial Cells/cytology , Animals , Cell Shape , Cells, Cultured , Hemodynamics , Ions/metabolism , Microscopy , Stress, Mechanical , Swine
9.
J R Soc Interface ; 8(60): 913-25, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21325316

ABSTRACT

Cardiovascular diseases are complex pathologies that include alterations of various cell functions at the levels of intact tissue, single cells and subcellular signalling compartments. Conventional techniques to study these processes are extremely divergent and rely on a combination of individual methods, which usually provide spatially and temporally limited information on single parameters of interest. This review describes scanning ion conductance microscopy (SICM) as a novel versatile technique capable of simultaneously reporting various structural and functional parameters at nanometre resolution in living cardiovascular cells at the level of the whole tissue, single cells and at the subcellular level, to investigate the mechanisms of cardiovascular disease. SICM is a multimodal imaging technology that allows concurrent and dynamic analysis of membrane morphology and various functional parameters (cell volume, membrane potentials, cellular contraction, single ion-channel currents and some parameters of intracellular signalling) in intact living cardiovascular cells and tissues with nanometre resolution at different levels of organization (tissue, cellular and subcellular levels). Using this technique, we showed that at the tissue level, cell orientation in the inner and outer aortic arch distinguishes atheroprone and atheroprotected regions. At the cellular level, heart failure leads to a pronounced loss of T-tubules in cardiac myocytes accompanied by a reduction in Z-groove ratio. We also demonstrated the capability of SICM to measure the entire cell volume as an index of cellular hypertrophy. This method can be further combined with fluorescence to simultaneously measure cardiomyocyte contraction and intracellular calcium transients or to map subcellular localization of membrane receptors coupled to cyclic adenosine monophosphate production. The SICM pipette can be used for patch-clamp recordings of membrane potential and single channel currents. In conclusion, SICM provides a highly informative multimodal imaging platform for functional analysis of the mechanisms of cardiovascular diseases, which should facilitate identification of novel therapeutic strategies.


Subject(s)
Aorta, Thoracic/physiology , Cardiovascular Diseases/pathology , Heart/physiology , Microscopy/methods , Myocytes, Cardiac/physiology , Animals , Aorta, Thoracic/ultrastructure , Humans , Microscopy/instrumentation , Myocytes, Cardiac/ultrastructure
10.
Arterioscler Thromb Vasc Biol ; 31(2): 384-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21127291

ABSTRACT

OBJECTIVE: The goal of this study was to examine the effect of chronic heterogeneous shear stress, applied using an orbital shaker, on endothelial cell morphology and the expression of cyclooxygenases 1 and 2. METHODS AND RESULTS: Porcine aortic endothelial cells were plated on fibronectin-coated Transwell plates. Cells were cultured for up to 7 days either under static conditions or on an orbital shaker that generated a wave of medium inducing shear stress over the cells. Cells were fixed and stained for the endothelial surface marker CD31 or cyclooxygenases 1 and 2. En face confocal microscopy and scanning ion conductance microscopy were used to show that endothelial cells were randomly oriented at the center of the well, aligned with shear stress nearer the periphery, and expressed cyclooxygenase-1 under all conditions. Lipopolysaccharide induced cyclooxygenase-2 and the production of 6-keto-prostaglandin F(1α) in all cells. CONCLUSIONS: Cyclooxygenase-1 is expressed in endothelial cells cultured under chronic shear stress of high or low directionality.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Stress, Mechanical , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cell Count , Cells, Cultured , Endothelium, Vascular/drug effects , Isoenzymes/metabolism , Lipopolysaccharides/pharmacology , Models, Animal , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...