Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.625
Filter
1.
Exp Biol Med (Maywood) ; 249: 10090, 2024.
Article in English | MEDLINE | ID: mdl-39143955

ABSTRACT

The intima, comprising the endothelium and the subendothelial matrix, plays a crucial role in atherosclerosis pathogenesis. The mechanical stress arising from disturbed blood flow (d-flow) and the stiffening of the arterial wall contributes to endothelial dysfunction. However, the specific impacts of these physical forces on the mechanical environment of the intima remain undetermined. Here, we investigated whether inhibiting collagen crosslinking could ameliorate the detrimental effects of persistent d-flow on the mechanical properties of the intima. Partial ligation of the left carotid artery (LCA) was performed in C57BL/6J mice, inducing d-flow. The right carotid artery (RCA) served as an internal control. Carotids were collected 2 days and 2 weeks after surgery to study acute and chronic effects of d-flow on the mechanical phenotype of the intima. The chronic effects of d-flow were decoupled from the ensuing arterial wall stiffening by administration of ß-aminopropionitrile (BAPN), an inhibitor of collagen crosslinking by lysyl oxidase (LOX) enzymes. Atomic force microscopy (AFM) was used to determine stiffness of the endothelium and the denuded subendothelial matrix in en face carotid preparations. The stiffness of human aortic endothelial cells (HAEC) cultured on soft and stiff hydrogels was also determined. Acute exposure to d-flow caused a slight decrease in endothelial stiffness in male mice but had no effect on the stiffness of the subendothelial matrix in either sex. Regardless of sex, the intact endothelium was softer than the subendothelial matrix. In contrast, exposure to chronic d-flow led to a substantial increase in the endothelial and subendothelial stiffness in both sexes. The effects of chronic d-flow were largely prevented by concurrent BAPN administration. In addition, HAEC displayed reduced stiffness when cultured on soft vs. stiff hydrogels. We conclude that chronic d-flow results in marked stiffening of the arterial intima, which can be effectively prevented by inhibition of collagen crosslinking.


Subject(s)
Carotid Arteries , Mice, Inbred C57BL , Vascular Stiffness , Animals , Male , Vascular Stiffness/drug effects , Mice , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Female , Tunica Intima/pathology , Tunica Intima/drug effects , Collagen/metabolism , Aminopropionitrile/pharmacology , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Microscopy, Atomic Force , Humans , Stress, Mechanical , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Endothelium, Vascular/metabolism
2.
Ulus Travma Acil Cerrahi Derg ; 30(8): 525-530, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39092970

ABSTRACT

BACKGROUND: Intimal hyperplasia is a normal adaptive feature of arteries in response to injuries, which include invasive vascular interventions. Its development limits the long-term success of bypass grafts. Various pharmacological agents have been successfully employed in experimental models to reduce the degree of intimal hyperplasia. In our study, we investigated the efficacy of dexamethasone in reducing intimal hyperplasia in rat abdominal aortas after partial transection and primary repair. METHODS: In this study, 20 Wistar Albino rats were randomly selected and divided into four groups to compare the effects of low- and high-dose dexamethasone on intima and media thickness compared to the control. Group A (n=5) was the control group, where only skin incision and laparotomy were performed. For Group B (n=5), a median laparotomy was performed, the abdominal aorta was partially transected, and repaired with an 8.0 prolene suture. Doses of 0.1 mg/kg and 0.2 mg/kg dexamethasone were administered in Group C (n=5) and Group D (n=5), respectively. After two weeks, all rats were euthanized, and the repaired abdominal aortas were excised and examined histopathologically. Intima and media thicknesses were measured using the 'Olympus AnalySIS 5' program (Olympus Corporation, Japan) after digital photos were taken. RESULTS: Based on the measurements, we demonstrated that after transection and repair of the abdominal aorta, the intima/media ratio was not significantly different between the low-dose dexamethasone and non-dexamethasone groups. The intima/media ratio was significantly lower in the high-dose dexamethasone group than in the non-dexamethasone and low-dose dexamethasone groups. CONCLUSION: After vascular interventions, dexamethasone treatment may reduce intimal hyperplasia and increase patency by providing vascular remodeling.


Subject(s)
Aorta, Abdominal , Dexamethasone , Hyperplasia , Rats, Wistar , Tunica Intima , Animals , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Aorta, Abdominal/surgery , Aorta, Abdominal/pathology , Rats , Hyperplasia/drug therapy , Hyperplasia/pathology , Hyperplasia/prevention & control , Tunica Intima/pathology , Tunica Intima/drug effects , Disease Models, Animal , Male
3.
Nanomedicine ; 61: 102763, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38897395

ABSTRACT

The primary challenge in percutaneous coronary interventions for vascular restenosis is the occurrence of restenosis, which is defined by the excessive proliferation of neointimal tissue. Herein, our research team suggests that exosomes obtained from PSC, when paired with quercetin (Q@PSC-E), successfully reduce neointimal hyperplasia in a Sprague-Dawley rat model. Furthermore, the physical properties of the synthesized Q@PSC-E were examined using UV-vis, DLS, and FT-IR characterization techniques. The rats were subjected to balloon injury (BI) utilizing a 2-Fr Fogarty arterial embolectomy balloon catheter. Intimal hyperplasia and the degree of VSMC proliferation were evaluated using histological analysis in the rat groups that received a dosage of Q@PSC-E at 30 mg/kg/d. Significantly, Q@PSC-E inhibited cell proliferation through a pathway that does not include lipoxygenase, as demonstrated by [3H] thymidine incorporation, MTT, and flow cytometry studies. Additionally, the data indicate that Q@PSC-E hinders cell proliferation by targeting particular events that promote cell growth, including the activation of Akt and NF-κB, disruption of cell-cycle progression and also obstructs the ERK signaling pathway.


Subject(s)
Cell Proliferation , Exosomes , Hyperplasia , NF-kappa B , Proto-Oncogene Proteins c-akt , Quercetin , Rats, Sprague-Dawley , Signal Transduction , Animals , NF-kappa B/metabolism , Rats , Proto-Oncogene Proteins c-akt/metabolism , Hyperplasia/pathology , Hyperplasia/drug therapy , Quercetin/pharmacology , Quercetin/chemistry , Exosomes/metabolism , Exosomes/drug effects , Signal Transduction/drug effects , Male , Cell Proliferation/drug effects , Carotid Artery Injuries/pathology , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/metabolism , Tunica Intima/pathology , Tunica Intima/drug effects , Tunica Intima/metabolism , MAP Kinase Signaling System/drug effects
4.
Thromb Res ; 238: 185-196, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729030

ABSTRACT

BACKGROUND: Plaque erosion, a type of coronary atherothrombosis, involves superficial injury to smooth muscle cell (SMC)-rich plaques. Elevated levels of coagulation factor VIII (FVIII) correlate with an increased ischemic heart disease risk. FVIII may contribute to thrombus formation on eroded plaques. AIMS: We aimed to elucidate the role of elevated FVIII in arterial thrombus formation within SMC-rich neointima in rabbits. METHODS AND RESULTS: We assessed the effect of recombinant human FVIII (rFVIII) on blood coagulation in vitro and platelet aggregation ex vivo. An SMC-rich neointima was induced through balloon injury to the unilateral femoral artery. Three weeks after the first balloon injury, superficial erosive injury and thrombus formation were initiated with a second balloon injury of the bilateral femoral arteries 45 min after the administration of rFVIII (100 IU/kg) or saline. The thrombus area and contents were histologically measured 15 min after the second balloon injury. rFVIII administration reduced the activated partial thromboplastin time and augmented botrocetin-induced, but not collagen- or adenosine 5'-diphosphate-induced, platelet aggregation. While rFVIII did not influence platelet-thrombus formation in normal intima, it increased thrombus formation on SMC-rich neointima post-superficial erosive injury. Enhanced immunopositivity for glycoprotein IIb/IIIa and fibrin was observed in rFVIII-administered SMC-rich neointima. Neutrophil count in the arterial thrombus on the SMC-rich neointima correlated positively with thrombus size in the control group, unlike the rFVIII group. CONCLUSIONS: Increased FVIII contributes to thrombus propagation within erosive SMC-rich neointima, highlighting FVIII's potential role in plaque erosion-related atherothrombosis.


Subject(s)
Factor VIII , Myocytes, Smooth Muscle , Neointima , Thrombosis , Rabbits , Animals , Neointima/pathology , Neointima/blood , Thrombosis/blood , Thrombosis/pathology , Male , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Tunica Intima/pathology , Tunica Intima/drug effects , Humans , Platelet Aggregation/drug effects , Femoral Artery/pathology , Femoral Artery/injuries
5.
Int J Mol Sci ; 25(9)2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38732029

ABSTRACT

Neointimal hyperplasia is the main cause of vascular graft failure in the medium term. Vitamin D receptor activation modulates the biology of vascular smooth muscle cells and has been reported to protect from neointimal hyperplasia following endothelial injury. However, the molecular mechanisms are poorly understood. We have now explored the impact of the selective vitamin D receptor activator, paricalcitol, on neointimal hyperplasia, following guidewire-induced endothelial cell injury in rats, and we have assessed the impact of paricalcitol or vehicle on the expression of key cell stress factors. Guidewire-induced endothelial cell injury caused neointimal hyperplasia and luminal stenosis and upregulated the expression of the growth factor growth/differentiation factor-15 (GDF-15), the cytokine receptor CD74, NFκB-inducing kinase (NIK, an upstream regulator of the proinflammatory transcription factor NFκB) and the chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2). Immunohistochemistry confirmed the increased expression of the cellular proteins CD74 and NIK. Paricalcitol (administered in doses of 750 ng/kg of body weight, every other day) had a non-significant impact on neointimal hyperplasia and luminal stenosis. However, it significantly decreased GDF-15, CD74, NIK and MCP-1/CCL2 mRNA expression, which in paricalcitol-injured arteries remained within the levels found in control vehicle sham arteries. In conclusion, paricalcitol had a dramatic effect, suppressing the stress response to guidewire-induced endothelial cell injury, despite a limited impact on neointimal hyperplasia and luminal stenosis. This observation identifies novel molecular targets of paricalcitol in the vascular system, whose differential expression cannot be justified as a consequence of improved tissue injury.


Subject(s)
Anti-Inflammatory Agents , Chemokine CCL2 , Ergocalciferols , Hyperplasia , Animals , Rats , Ergocalciferols/pharmacology , Male , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Anti-Inflammatory Agents/pharmacology , Neointima/metabolism , Neointima/pathology , Neointima/drug therapy , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Tunica Intima/pathology , Tunica Intima/drug effects , Tunica Intima/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Histocompatibility Antigens Class II
6.
Hypertens Res ; 47(7): 1908-1924, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750218

ABSTRACT

Phenotypic shift of vascular smooth muscle cells (VSMCs) plays a key role in intimal hyperplasia, especially in patients with diabetes mellitus (DM). This study aimed to investigate the role of dynamin-related protein 1 (DRP1) in mitochondrial fission-mediated VSMC phenotypic shift and to clarify whether DRP1 is the therapeutic target of isoliquiritigenin (ISL). Wire injury of carotid artery or platelet-derived growth factor treatment was performed in DM mice or high-glucose cultured human aortic smooth muscle cells (HASMCs), respectively. The effects of DRP1 silencing on DM-induced intimal hyperplasia were investigated both in vivo and in vitro. Phenotypic shift of HASMCs was evaluated by detection of reactive oxygen species (ROS) generation, cell viability, and related protein expressions. The effects of ISL on DM-induced intimal hyperplasia were evaluated both in vivo and in vitro. DRP1 silencing and ISL treatment attenuated DM-induced intimal hyperplasia with reduced ROS generation, cell viability, and VSMC dedifferentiation. The GTPase domain of DRP1 protein played a critical role in mitochondrial fission in DM-induced VSMC phenotypic shift. Cellular experiments showed that ISL inhibited mitochondrial fission and reduced the GTPase activity of DRP1, which was achieved by the directly binding to K216 of the DRP1 GTPase domain. ISL attenuated mouse intimal hyperplasia by reducing GTPase activity of DRP1 and inhibiting mitochondrial fission in vivo. In conclusion, increased GTPase activity of DRP1 aggregated DM-induced intimal hyperplasia by increasing mitochondrial fission-mediated VSMC phenotypic shift. ISL attenuated mouse intimal hyperplasia by reducing DRP1 GTPase activity and inhibiting mitochondrial fission of VSMCs.


Subject(s)
Chalcones , Dynamins , Hyperplasia , Mitochondrial Dynamics , Animals , Mitochondrial Dynamics/drug effects , Dynamins/metabolism , Chalcones/pharmacology , Chalcones/therapeutic use , Mice , Humans , Male , Diabetes Mellitus, Experimental/drug therapy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Reactive Oxygen Species/metabolism , Myocytes, Smooth Muscle/drug effects , Cells, Cultured , Mice, Inbred C57BL , Tunica Intima/drug effects , Tunica Intima/pathology , Tunica Intima/metabolism
7.
Morphologie ; 108(362): 100779, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38608628

ABSTRACT

BACKGROUND: One of the most recent hormones to be identified and isolated is irisin, extracted from mouse skeletal muscle in 2012. Irisin has been proven to alter blood pressure, which has an impact on blood vessels, enhance endothelial functions, and prevent injury to endothelial cells. The current study aimed to study the effect of irisin on the ultrastructure of the rat thoracic aorta using the transmission electron microscope (TEM). MATERIALS AND METHODS: Twenty female rats were recruited for this study and divided into a control group (non-injected), and four experimental groups (injected groups) each consisting of 4 rats. The experimental groups were injected intraperitoneally with different doses of irisin (250ng/mL, 500ng/mL, 1000ng/mL, and 2000ng/mL) twice a week for 4weeks. Then, the descending thoracic aorta of all experimental rats were resected and proceeded with imaging. RESULTS: The results of this study showed a change in the thickness of the tunica intima, internal elastic lamina, elastic lamellae, and external elastic lamina concerning increasing injected irisin concentration. While there was a significant increase in the thickness of tunica media (P<0.0001) and smooth muscle cells (P<0.05). Also, the results showed a significant increase in the number of elastic lamellae in the tunica media (P<0.0001). CONCLUSION: Irisin had a major impact on the elasticity of the rat thoracic aorta wall, suggesting that it influences the growth factors of the wall and activates smooth muscle cells in addition to endothelial cells.


Subject(s)
Aorta, Thoracic , Fibronectins , Microscopy, Electron, Transmission , Animals , Fibronectins/pharmacology , Aorta, Thoracic/drug effects , Aorta, Thoracic/ultrastructure , Rats , Female , Tunica Intima/ultrastructure , Tunica Intima/drug effects , Rats, Sprague-Dawley , Tunica Media/drug effects , Tunica Media/ultrastructure
8.
Eur J Vasc Endovasc Surg ; 63(2): 336-346, 2022 02.
Article in English | MEDLINE | ID: mdl-34916111

ABSTRACT

OBJECTIVE: Hypertension is a major risk factor for intimal hyperplasia (IH) and re-stenosis following vascular and endovascular interventions. Preclinical studies suggest that hydrogen sulphide (H2S), an endogenous gasotransmitter, limits re-stenosis. While there is no clinically available pure H2S releasing compound, the sulfhydryl containing angiotensin converting enzyme inhibitor zofenopril is a source of H2S. Here, it was hypothesised that zofenopril, due to H2S release, would be superior to other non-sulfhydryl containing angiotensin converting enzyme inhibitors (ACEi) in reducing intimal hyperplasia. METHODS: Spontaneously hypertensive male Cx40 deleted mice (Cx40-/-) or wild type (WT) littermates were randomly treated with enalapril 20 mg or zofenopril 30 mg. Discarded human vein segments and primary human smooth muscle cells (SMCs) were treated with the active compound enalaprilat or zofenoprilat. IH was evaluated in mice 28 days after focal carotid artery stenosis surgery and in human vein segments cultured for seven days ex vivo. Human primary smooth muscle cell (SMC) proliferation and migration were studied in vitro. RESULTS: Compared with control animals (intima/media thickness 2.3 ± 0.33 µm), enalapril reduced IH in Cx40-/- hypertensive mice by 30% (1.7 ± 0.35 µm; p = .037), while zofenopril abrogated IH (0.4 ± 0.16 µm; p < .002 vs. control and p > .99 vs. sham operated Cx40-/- mice). In WT normotensive mice, enalapril had no effect (0.9665 ± 0.2 µm in control vs. 1.140 ± 0.27 µm; p > .99), while zofenopril also abrogated IH (0.1623 ± 0.07 µm; p < .008 vs. control and p > .99 vs. sham operated WT mice). Zofenoprilat, but not enalaprilat, also prevented IH in human vein segments ex vivo. The effect of zofenopril on carotid and SMCs correlated with reduced SMC proliferation and migration. Zofenoprilat inhibited the mitogen activated protein kinase and mammalian target of rapamycin pathways in SMCs and human vein segments. CONCLUSION: Zofenopril provides extra beneficial effects compared with non-sulfhydryl ACEi in reducing SMC proliferation and re-stenosis, even in normotensive animals. These findings may hold broad clinical implications for patients suffering from vascular occlusive diseases and hypertension.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Captopril/analogs & derivatives , Carotid Stenosis/drug therapy , Hypertension/complications , Tunica Intima/pathology , Animals , Blood Pressure/drug effects , Captopril/administration & dosage , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Cells, Cultured , Disease Models, Animal , Humans , Hydrogen Sulfide/metabolism , Hyperplasia/drug therapy , Hyperplasia/pathology , Hypertension/drug therapy , Male , Mice , Myocytes, Smooth Muscle , Organ Culture Techniques , Primary Cell Culture , Tunica Intima/drug effects , Veins/drug effects , Veins/pathology
9.
J Nanobiotechnology ; 19(1): 374, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34789284

ABSTRACT

BACKGROUND: Intimal hyperplasia caused by vascular injury is an important pathological process of many vascular diseases, especially occlusive vascular disease. In recent years, Nano-drug delivery system has attracted a wide attention as a novel treatment strategy, but there are still some challenges such as high clearance rate and insufficient targeting. RESULTS: In this study, we report a biomimetic ROS-responsive MM@PCM/RAP nanoparticle coated with macrophage membrane. The macrophage membrane with the innate "homing" capacity can superiorly regulate the recruitment of MM@PCM/RAP to inflammatory lesion to enhance target efficacy, and can also disguise MM@PCM/RAP nanoparticle as the autologous cell to avoid clearance by the immune system. In addition, MM@PCM/RAP can effectively improve the solubility of rapamycin and respond to the high concentration level of ROS accumulated in pathological lesion for controlling local cargo release, thereby increasing drug availability and reducing toxic side effects. CONCLUSIONS: Our findings validate that the rational design, biomimetic nanoparticles MM@PCM/RAP, can effectively inhibit the pathological process of intimal injury with excellent biocompatibility.


Subject(s)
Hyperplasia/metabolism , Macrophages/cytology , Nanoparticle Drug Delivery System , Reactive Oxygen Species/metabolism , Tunica Intima , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Cell Membrane/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Male , Mice , Mice, Inbred C57BL , Nanoparticle Drug Delivery System/chemistry , Nanoparticle Drug Delivery System/metabolism , Sirolimus/chemistry , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Tunica Intima/drug effects , Tunica Intima/metabolism , Zebrafish
10.
Respir Res ; 22(1): 281, 2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34717626

ABSTRACT

BACKGROUND: This study examined whether BI113823, a novel selective kinin B1 receptor antagonist can reverse established pulmonary arterial hypertension (PAH), prevent right heart failure and death, which is critical for clinical translation. METHODS: Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Three weeks later, when PAH was well established, the rats received daily treatment of BI113823 or vehicle for 3 weeks. RESULTS: Treatment with BI113823 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values of mean pulmonary arterial pressure (mPAP). BI113823 therapy reversed pulmonary vascular remodeling, pulmonary arterial neointimal formation, and heart and lung fibrosis, reduced right ventricular pressure, right heart hypertrophy, improved cardiac output, and prevented right heart failure and death. Treatment with BI113823 reduced TNF-α and IL-1ß, and macrophages recruitment in bronchoalveolar lavage, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen (PCNA) in the perivascular areas, and reduced expression of iNOS, B1 receptors, matrix metalloproteinase (MMP)-2 and MMP-9 proteins, and the phosphorylation of ERK1/2 and AKT in lung. Treatment with BI113823 reduced mRNA expression of ANP, BNP, ßMHC, CGTF, collange-I and IV in right heart, compared to vehicle treated controls. In human monocytes cultures, BI113823 reduced LPS-induced TNF-α production, MMP-2 and MMP-9 expression, and reduced TNF-α-induced monocyte migration. CONCLUSIONS: We conclude that BI113823 reverses preexisting severe experimental pulmonary hypertension via inhibition of macrophage infiltration, cytokine production, as well as down regulation of matrix metalloproteinase proteins.


Subject(s)
Kinins/antagonists & inhibitors , Neointima/pathology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/pathology , Tunica Intima/pathology , Vascular Remodeling/drug effects , Animals , Disease Models, Animal , Humans , Male , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Tunica Intima/drug effects
11.
Rheumatology (Oxford) ; 60(11): 5052-5059, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34117737

ABSTRACT

OBJECTIVES: To characterize the effect of ultra-short glucocorticoids followed by Tocilizumab monotherapy on the intima-media thickness (IMT) in GCA. METHODS: Eighteen GCA patients received 500 mg for 3 consecutive days (total of 1500mg) i.v. methylprednisolone on days 0-2, followed by i.v. Tocilizumab (8 mg/kg) on day 3 and thereafter weekly s.c. Tocilizumab injections (162 mg) over 52 weeks. US of temporal (TAs), axillary (AAs) and subclavian (SAs) arteries was performed at baseline, on days 2-3, and at weeks 4, 8, 12, 24 and 52. The largest IMT of all segments and IMT at landmarks of AA/SA were recorded. IMT was scaled by mean normal values and averaged. Each segment was classified according to diagnostic cut-offs. RESULTS: Of the 18 GCA patients, 16 patients had TA and 6 had extracranial large artery involvement. The IMT showed a sharp decline on day 2/3 in the TAs and AAs/SAs. In TAs, this was followed by an increase to baseline levels at week 4 and a subsequent slow decrease, which was paralleled by decreasing symptoms and achievement of clinical remission. The AAs/SAs showed a new signal of vasculitis at week 4 in three patients, with an IMT increase up to week 8. CONCLUSION: Glucocorticoid pulse therapy induced a transient decrease of the IMT in TAs and AAs/SAs. Tocilizumab monotherapy resulted in a slow and steady decrease in IMT of the TAs and a smaller and delayed effect on the AAs/SAs. The data strongly support a remission-inducing effect of Tocilizumab and argue the case for US having an important role in monitoring disease activity in GCA. TRIAL REGISTRATION: ClinicalTrials.gov, www.clinicaltrials.gov, NCT03745586.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Giant Cell Arteritis/diagnostic imaging , Giant Cell Arteritis/drug therapy , Glucocorticoids/therapeutic use , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Arteries/diagnostic imaging , Arteries/drug effects , Female , Glucocorticoids/pharmacology , Humans , Male , Proof of Concept Study , Tunica Intima/diagnostic imaging , Tunica Intima/drug effects , Ultrasonography
12.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467058

ABSTRACT

Vascular dysfunction in cardiovascular diseases includes vasomotor response impairments, endothelial cells (ECs) activation, and smooth muscle cells (SMCs) proliferation and migration to the intima. This results in intimal hyperplasia and vessel failure. We previously reported that activation of the P2Y11 receptor (P2Y11R) in human dendritic cells, cardiofibroblasts and cardiomyocytes was protective against hypoxia/reoxygenation (HR) lesions. In this study, we investigated the role of P2Y11R signaling in vascular dysfunction. P2Y11R activity was modulated using its pharmacological agonist NF546 and antagonist NF340. Rat aortic rings were exposed to angiotensin II (AngII) and evaluated for their vasomotor response. The P2Y11R agonist NF546 reduced AngII-induced vascular dysfunction by promoting EC-dependent vasorelaxation, through an increased nitric oxide (NO) bioavailability and reduced AngII-induced H2O2 release; these effects were prevented by the use of the P2Y11R antagonist NF340. Human vascular SMCs and ECs were subjected to AngII or H/R simulation in vitro. P2Y11R agonist modulated vasoactive factors in human ECs, that is, endothelial nitric oxide synthase (eNOS) and endothelin-1, reduced SMC proliferation and prevented the switch towards a synthetic phenotype. H/R and AngII increased ECs secretome-induced SMC proliferation, an effect prevented by P2Y11R activation. Thus, our data suggest that P2Y11R activation may protect blood vessels from HR-/AngII-induced injury and reduce vascular dysfunctions. These results open the way for new vasculoprotective interventions.


Subject(s)
Diphosphonates/pharmacology , Naphthalenesulfonates/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2/metabolism , Reperfusion Injury/metabolism , Tunica Intima/pathology , Angiotensin II/toxicity , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Diphosphonates/therapeutic use , Endothelin-1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyperplasia/prevention & control , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Naphthalenesulfonates/therapeutic use , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Purinergic P2 Receptor Agonists/therapeutic use , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Tunica Intima/drug effects , Tunica Intima/metabolism , Vasodilation , Water/metabolism
13.
Exp Biol Med (Maywood) ; 246(2): 226-239, 2021 01.
Article in English | MEDLINE | ID: mdl-32996350

ABSTRACT

Restenosis after angioplasty of peripheral arteries is a clinical problem involving oxidative stress. Hydrogen sulfide (H2S) participates in oxidative stress regulation and activates nuclear factor erythroid 2-related factor 2 (Nrf2). This study investigated the effect of H2S and Nrf2 on restenosis-induced arterial injury. Using an in vivo rat model of restenosis, we investigated whether H2S inhibits restenosis after percutaneous transluminal angioplasty (PTA) and the oxidative stress-related mechanisms implicated therein. The involvement of Nrf2 was explored using Nrf2-shRNA. Neointimal formation and the deposition of elastic fibers were assessed histologically. Inflammatory cytokine secretion and the expression of proteins associated with oxidative stress and inflammation were evaluated. The artery of rats subjected to restenosis showed increased arterial intimal thickness, with prominent elastic fiber deposition. Sodium hydrosulfide (NaHS), an H2S donor, counteracted these changes in vivo. Restenosis caused a decrease in anti-oxidative stress signaling. This phenomenon was inhibited by NaHS, but Nrf2-shRNA counteracted the effects of NaHS. In terms of inflammation, inflammatory cytokines were upregulated, whereas NaHS suppressed the induced inflammatory reaction. Similarly, Nrf2 downregulation blocked the effect of NaHS. In vitro studies using aortic endothelial and vascular smooth muscle cells isolated from experimental animals showed consistent results as those of in vivo studies, and the participation of the nuclear factor-kappa B signaling pathway was demonstrated. Collectively, H2S played a role in regulating post-PTA restenosis by alleviating oxidative stress, modulating anti-oxidant defense, and targeting Nrf2-related pathways via nuclear factor-kappa B signaling.


Subject(s)
Angioplasty/adverse effects , Coronary Restenosis/etiology , Coronary Restenosis/pathology , Hydrogen Sulfide/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Coronary Restenosis/metabolism , Hyperplasia , Inflammation/pathology , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxidation-Reduction , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tunica Intima/drug effects , Tunica Intima/pathology
15.
Theranostics ; 10(17): 7787-7811, 2020.
Article in English | MEDLINE | ID: mdl-32685020

ABSTRACT

Objective: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are essential for vascular remodeling. Natural compounds with diterpene chinone or phenolic acid structure from Salvia miltiorrhiza, an eminent medicinal herb widely used to treat cardiovascular diseases in China, can effectively attenuate vascular remodeling induced by vascular injury. However, it remains unknown whether Salvia miltiorrhiza-derived miRNAs can protect VSMCs from injury by environmental stimuli. Here, we explored the role and underlying mechanisms of Salvia miltiorrhiza-derived Sal-miR-1 and 3 in the regulation of VSMC migration and monocyte adhesion to VSMCs induced by thrombin. Methods: A mouse model for intimal hyperplasia was established by the ligation of carotid artery and the injured carotid arteries were in situ-transfected with Sal-miR-1 and 3 using F-127 pluronic gel. The vascular protective effects of Sal-miR-1 and 3 were assessed via analysis of intimal hyperplasia with pathological morphology. VSMC migration and adhesion were analyzed by the wound healing, transwell membrane assays, and time-lapse imaging experiment. Using loss- and gain-of-function approaches, Sal-miR-1 and 3 regulation of OTUD7B/KLF4/NMHC IIA axis was investigated by using luciferase assay, co-immunoprecipitation, chromatin immunoprecipitation, western blotting, etc. Results:Salvia miltiorrhiza-derived Sal-miR-1 and 3 can enter the mouse body after intragastric administration, and significantly suppress intimal hyperplasia induced by carotid artery ligation. In cultured VSMCs, these two miRNAs inhibit thrombin-induced the migration of VSMCs and monocyte adhesion to VSMCs. Mechanistically, Sal-miR-1 and 3 abrogate OTUD7B upregulation by thrombin via binding to the different sites of the OTUD7B 3'UTR. Most importantly, OTUD7B downregulation by Sal-miR-1 and 3 attenuates KLF4 protein levels via decreasing its deubiquitylation, whereas decreased KLF4 relieves its repression of transcription of NMHC IIA gene and thus increases NMHC IIA expression levels. Further, increased NMHC IIA represses VSMC migration and monocyte adhesion to VSMCs via maintaining the contractile phenotype of VSMCs. Conclusions: Our studies not only found the novel bioactive components from Salvia miltiorrhiza but also clarified the molecular mechanism underlying Sal-miR-1 and 3 inhibition of VSMC migration and monocyte adhesion to VSMCs. These results add important knowledge to the pharmacological actions and bioactive components of Salvia miltiorrhiza. Sal-miR-1 and 3-regulated OTUD7B/KLF4/NMHC IIA axis may represent a therapeutic target for vascular remodeling.


Subject(s)
MicroRNAs/pharmacology , RNA, Plant/pharmacology , Salvia miltiorrhiza/genetics , Tunica Intima/pathology , Vascular Remodeling/drug effects , Animals , Carotid Arteries/cytology , Carotid Arteries/pathology , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation , Endopeptidases/metabolism , Humans , Hyperplasia/drug therapy , Hyperplasia/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , MicroRNAs/therapeutic use , Monocytes/drug effects , Monocytes/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Myosin Heavy Chains/metabolism , RNA, Plant/therapeutic use , Signal Transduction/drug effects , Tunica Intima/drug effects
16.
Transl Res ; 224: 40-54, 2020 10.
Article in English | MEDLINE | ID: mdl-32522668

ABSTRACT

The modulation of voltage-gated K+ (Kv) channels, involved in cell proliferation, arises as a potential therapeutic approach for the prevention of intimal hyperplasia present in in-stent restenosis (ISR) and allograft vasculopathy (AV). We studied the effect of PAP-1, a selective blocker of Kv1.3 channels, on development of intimal hyperplasia in vitro and in vivo in 2 porcine models of vascular injury. In vitro phenotypic modulation of VSMCs was associated to an increased functional expression of Kv1.3 channels, and only selective Kv1.3 channel blockers were able to inhibit porcine VSMC proliferation. The therapeutic potential of PAP-1 was then evaluated in vivo in swine models of ISR and AV. At 15-days follow-up, morphometric analysis demonstrated a substantial reduction of luminal stenosis in the allografts treated with PAP-1 (autograft 2.72 ± 1.79 vs allograft 10.32 ± 1.92 vs allograft + polymer 13.54 ± 8.59 vs allograft + polymer + PAP-1 3.06 ± 1.08 % of luminal stenosis; P = 0.006) in the swine model of femoral artery transplant. In the pig model of coronary ISR, using a prototype of PAP-1-eluting stent, no differences were observed regarding % of stenosis compared to control stents (31 ± 13 % vs 37 ± 18%, respectively; P = 0.372) at 28-days follow-up. PAP-1 treatment was safe and did not impair vascular healing in terms of delayed endothelialization, inflammation or thrombosis. However, an incomplete release of PAP-1 from stents was documented. We conclude that the use of selective Kv1.3 blockers represents a promising therapeutic approach for the prevention of intimal hyperplasia in AV, although further studies to improve their delivery method are needed to elucidate its potential in ISR.


Subject(s)
Kv1.3 Potassium Channel/antagonists & inhibitors , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Potassium Channel Blockers/pharmacology , Tunica Intima/pathology , Allografts/drug effects , Animals , Cell Proliferation/drug effects , Coronary Restenosis/pathology , Coronary Vessels/drug effects , Coronary Vessels/injuries , Coronary Vessels/pathology , Disease Models, Animal , Female , Femoral Artery/drug effects , Femoral Artery/pathology , Gene Expression Regulation/drug effects , Hyperplasia , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Models, Biological , Myocytes, Smooth Muscle/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stents , Swine , Tunica Intima/drug effects
17.
Cell Cycle ; 19(8): 918-932, 2020 04.
Article in English | MEDLINE | ID: mdl-32202193

ABSTRACT

Excessive migration and proliferation of vascular smooth muscle cells (VSMCs) are critical cellular events that lead to intimal hyperplasia in atherosclerosis and restenosis. In this study, we investigated the protective effects of ursodeoxycholic acid (UDCA) on intimal hyperplasia and VSMC proliferation and migration, and the underlying mechanisms by which these events occur. A rat unilateral carotid artery was ligated to induce vascular injury and the microRNA (miRNA) expression profiles were determined using miRNA microarray analysis. We observed that UDCA significantly reduced the degree of intimal hyperplasia and induced miR-21 dysregulation. Restoration of miR-21 by agomir-miR-21 reversed the protective effects of UDCA on intimal hyperplasia and proliferation in vivo. In vitro, UDCA suppressed PDGF-BB-induced VSMC proliferation, invasion and migration in a dose-dependent manner, whereas the suppressive effect of UDCA was abrogated by overexpression of miR-21 in PDGF-BB-incubated VSMCs. Furthermore, we identified that miR-21 in VSMCs targeted the phosphatase and tensin homolog (PTEN), a tumor suppressor gene, negatively modulated the AKT/mTOR pathway. More importantly, we observed that that UDCA suppressed AKT/mTOR signaling pathway in the carotid artery injury model, whereas this pathway was reactivated by overexpression of miR-21. Taken together, our findings indicated that UDCA inhibited intimal hyperplasia and VSMCs excessive migration and proliferation via blocking miR-21/PTEN/AKT/mTOR signaling pathway, which suggests that UDCA may be a promising candidate for the therapy of atherosclerosis.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Hyperplasia/drug therapy , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tunica Intima/pathology , Ursodeoxycholic Acid/administration & dosage , Vascular System Injuries/drug therapy , Vascular System Injuries/metabolism , Animals , Antagomirs/administration & dosage , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Male , MicroRNAs/genetics , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , PTEN Phosphohydrolase/genetics , Rats , Rats, Sprague-Dawley , Transfection , Tunica Intima/drug effects , Vascular System Injuries/pathology
18.
J Nutr Sci Vitaminol (Tokyo) ; 66(1): 75-81, 2020.
Article in English | MEDLINE | ID: mdl-32115457

ABSTRACT

Abdominal aortic aneurysm (AAA) is a vascular disease characterized by weakening of vascular walls and progressive dilation of the abdominal aorta. Nicotine, the main component of tobacco, is reportedly associated with the development and rupture of AAA. It is desirable to attenuate the destructive effect of nicotine on vascular walls, using dietary food components. However, effective methods for preventing AAA progression using dietary food components remain unestablished. This study focuses on proanthocyanidins, well known for their potent antioxidant activity. We speculated that proanthocyanidins can suppress nicotine-induced weakening of vascular walls. To estimate the effect of black soybean seed coat extract (BSSCE), rich in proanthocyanidins, on nicotine-induced weakening of the aortic wall, mice were divided into four groups: the control diet and distilled water group (named C), BSSCE solution diet and distilled water group (named B), control diet and 0.5 mg/mL nicotine solution group (named CN), and BSSCE solution diet and 0.5 mg/mL nicotine solution group (named BN). Nicotine-induced degradation of elastin and collagen fibers were significantly suppressed in BN group. The positive areas for matrix metalloproteinase (MMP)-2 and oxidative stress in BN group were significantly decreased compared to those in CN group. These results suggest that proanthocyanidins-rich BSSCE can prevent the weakening of the aortic wall via inhibiting MMP-2 upregulation.


Subject(s)
Aorta , Glycine max/chemistry , Matrix Metalloproteinase 2/metabolism , Nicotine/adverse effects , Plant Extracts/pharmacology , Adventitia/drug effects , Adventitia/metabolism , Adventitia/pathology , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/physiopathology , Aortic Aneurysm, Abdominal , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Plant Extracts/chemistry , Proanthocyanidins/chemistry , Proanthocyanidins/pharmacology , Seeds/chemistry , Tobacco Smoke Pollution , Tunica Intima/drug effects , Tunica Intima/metabolism , Tunica Intima/pathology
19.
J Cardiothorac Surg ; 15(1): 34, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32041642

ABSTRACT

BACKGROUND: This study aims to compare the effects of storage solutions commonly used in coronary artery bypass grafting on the vascular reactivity in vein graft interposed in arterial position in syngeneic rats. METHODS: Twenty-seven male Lewis rats were sacrified to sample a vein graft implanted 6 weeks ago into abdominal aorta position. The vein grafts were inferior venae cavae initially pretreated with heparinized saline solution (HS) or autologous heparinized blood (AHB) or our referent solution, GALA. The endothelial functionality, the in situ Reactive Oxygen Species (ROS) levels and the histological characteristics were conducted from segments of arterialized vein graft. RESULTS: At 6 weeks, graft thrombosis occurred respectively in 22% of AHB group, 62.5% in the HS group and 82.5% in the GALA group. In each group, significative intimal hyperplasia was observed. After 6 weeks, an endothelium-remodeling layer associated with an increase of wall thickness was observed in each group. Endothelium-dependent tone was reduced in the vein graft regardless of the group. No difference was observed concerning the ROS in vein graft between the different groups. In distal aortic sections, ROS levels were increased in HS and GALA groups. CONCLUSIONS: Storage solutions used in this experimental model of vein graft implanted in arterial position cause graft injury and a complete disappearance of vascular reactivity. GALA solution did not reduce intimal risk hyperplasia when the vein graft was exposed to arterial flow in a rat model.


Subject(s)
Aorta, Abdominal/surgery , Coronary Artery Bypass , Endothelium, Vascular/drug effects , Organ Preservation Solutions/pharmacology , Tunica Intima/pathology , Vena Cava, Inferior/transplantation , Animals , Anticoagulants/administration & dosage , Anticoagulants/therapeutic use , Blood , Disease Models, Animal , Endothelium, Vascular/pathology , Heparin/administration & dosage , Heparin/therapeutic use , Hyperplasia , Male , Organ Preservation Solutions/administration & dosage , Organ Preservation Solutions/therapeutic use , Rats , Rats, Inbred Lew , Reactive Oxygen Species/analysis , Saline Solution/administration & dosage , Saline Solution/therapeutic use , Tunica Intima/drug effects , Vena Cava, Inferior/drug effects
20.
Biomed Pharmacother ; 124: 109935, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31986407

ABSTRACT

Intimal hyperplasia, the key event of arterial restenosis, is a result of vascular smooth muscle cell (VSMC) proliferation and migration. Previous studies have demonstrated that total Panax notoginseng saponin (TPNS) represses intimal hyperplasia and inhibits the proliferation of VSMCs following balloon injury. However, the underlying roles of TPNS in intimal hyperplasia remain unclear. In this study, we first found that TPNS inhibited the intimal hyperplasia and reversed the reduced m6A quantity in balloon catheter-injured rat carotid artery. Then, we measured the expression profiles of m6A "writers" (i.e., methyltransferase like 3 (METTL3), methyltransferase like 14 (METTL14), and WT1 associated protein (WTAP)) and "erasers" (i.e., FTO alpha-ketoglutarate dependent dioxygenase (FTO) and alkB homolog 5, RNA demethylase (ALKBH5)) in vivo and found that TPNS up-regulated the reduced the WTAP expression in balloon catheter-injured rat carotid artery. Furthermore, we illustrated that TPNS inhibited the viability, proliferation, and migration potential of VSMCs via promotion of WTAP expression and suppression of WTAP restored the TPNS-induced inhibition of cell viability, proliferation and migration potential of VSMCs. In addition, we found that p16 was up-regulated in VSMCs treated with TPNS and repression of p16 restored the TPNS-induced inhibition of cell viability, proliferation and migration potential of VSMCs. Finally, we elucidated that, mechanistically, WTAP exerted its role by regulating p16 via m6A modification. Collectively, our results reveal the WTAP-p16 signaling axis and highlight the critical roles of m6A modification in intimal hyperplasia. Thus, this study provided a potential biomarker for the assessment of intimal hyperplasia risk following angioplasty as well as a novel therapeutic target for this disease.


Subject(s)
Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Panax notoginseng/chemistry , Saponins/pharmacology , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Hyperplasia/drug therapy , Male , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Saponins/isolation & purification , Signal Transduction/drug effects , Tunica Intima/drug effects , Tunica Intima/pathology , WT1 Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL