1
|
Basaran K, Datli A, Sagir M, Sar M, Bilgi E. Blunt Trauma Induced Closed Femoral Bone Fracture in a Rat Model: Are Vessels Safe to Use for Microsurgery? Further Insight into the Zone of Injury Concept. J Reconstr Microsurg 2024. [PMID: 38782027 DOI: 10.1055/a-2332-0263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND The study aims to investigate the zone of injury for major vessels after high-velocity traumas, as it is unclear whether avoiding vascular structures is necessary during microvascular anastomosis or how long it takes for them to be used again. METHODS This study uses Doppler ultrasonography and a rat model to evaluate the histopathological changes and flow velocity of major vessels in the zone of injury after high-velocity trauma with closed femoral bone fracture. Osteosynthesis was performed using an intramedullary wire. Samples were collected from day 3 and week 3. The unaffected contralateral side is used as control. RESULTS Results from arterial and venous flow assessments showed no evidence of ischemia in the extremities. Both arteries and veins were patent in both intervals and on the control side. The evaluation of the vessels showed arterial injury with a slightly reduced arterial flow on day 3 and week 3. The venous flow was slightly reduced on day 3 but not on week 3. Statistically, arterial endothelial injury was higher on day 3 than on week 3 (p = 0.006). Media inflammation was also higher on day 3 (p = 0.06). Arterial endothelization distribution was higher in week 3 (p = 0.006). No significant differences were found in arterial media irregularity, necrosis, platelet aggregation, bleeding, and wall rupture. Venous samples showed no significant differences in any parameter (p < 0.05). CONCLUSION High-velocity trauma increases the risk of thrombosis in vessels. Intravascular repair can start on day 2 and continue till week 3 with significant endothelization. Although physiologic findings do not alter arterial or venous flow, histologic findings support vessel injuries leading to potential complications. Microsurgery should be considered out of the injury zone until adequate vessel healing is achieved.
Collapse
Affiliation(s)
- Karaca Basaran
- Uskudar University, Plastic, Reconstructive and Aesthetic Surgery Department, Istanbul, Turkiye
| | - Asli Datli
- Istinye University, Plastic, Reconstructive and Aesthetic Surgery Department, Istanbul, Turkiye
| | - Mehmet Sagir
- Private Acibadem Hospital, Plastic, Reconstructive and Aesthetic Surgery Department, Istanbul, Turkiye
| | | | - Esra Bilgi
- Health Science University, Radiology Department, Istanbul, Turkiye
| |
Collapse
|
2
|
Bernad SI, Socoliuc V, Craciunescu I, Turcu R, Bernad ES. Field-Induced Agglomerations of Polyethylene-Glycol-Functionalized Nanoclusters: Rheological Behaviour and Optical Microscopy. Pharmaceutics 2023; 15:2612. [PMID: 38004590 PMCID: PMC10675764 DOI: 10.3390/pharmaceutics15112612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
This research aims to investigate the agglomeration processes of magnetoresponsive functionalized nanocluster suspensions in a magnetic field, as well as how these structures impact the behaviour of these suspensions in biomedical applications. The synthesis, shape, colloidal stability, and magnetic characteristics of PEG-functionalized nanoclusters are described in this paper. Experiments using TEM, XPS, dynamic light scattering (DLS), VSM, and optical microscopy were performed to study chain-like agglomeration production and its influence on colloidal behaviour in physiologically relevant suspensions. The applied magnetic field aligns the magnetic moments of the nanoclusters. It provides an attraction between neighbouring particles, resulting in the formation of chains, linear aggregates, or agglomerates of clusters aligned along the applied field direction. Optical microscopy has been used to observe the creation of these aligned linear formations. The design of chain-like structures can cause considerable changes in the characteristics of ferrofluids, ranging from rheological differences to colloidal stability changes.
Collapse
Affiliation(s)
- Sandor I. Bernad
- Centre for Fundamental and Advanced Technical Research, Romanian Academy—Timisoara Branch, Mihai Viteazul Str. 24, RO-300223 Timisoara, Romania;
| | - Vlad Socoliuc
- Centre for Fundamental and Advanced Technical Research, Romanian Academy—Timisoara Branch, Mihai Viteazul Str. 24, RO-300223 Timisoara, Romania;
| | - Izabell Craciunescu
- National Institute for Research and Development of Isotopic and Molecular Technologies (INCDTIM), Donat Str. 67-103, RO-400293 Cluj-Napoca, Romania; (I.C.); (R.T.)
| | - Rodica Turcu
- National Institute for Research and Development of Isotopic and Molecular Technologies (INCDTIM), Donat Str. 67-103, RO-400293 Cluj-Napoca, Romania; (I.C.); (R.T.)
| | - Elena S. Bernad
- Department of Obstetrics and Gynecology, Faculty of General Medicine, University of Medicine and Pharmacy “Victor Babes” Timisoara, P-ta Eftimie Murgu 2, RO-300041 Timisoara, Romania;
| |
Collapse
|
3
|
Kler A, Khambalia H, Pondor Z, Donne R, Giuffrida G, Augustine T, Campbell B, van Dellen D. Smoking is associated with a higher complication and failure rate in arteriovenous grafts for haemodialysis: A multi-centre experience. J Vasc Access 2023; 24:747-753. [PMID: 34727765 DOI: 10.1177/11297298211054634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Arteriovenous grafts (AVG) for haemodialysis (HD) access are recommended as a second line modality due to higher morbidity and mortality rates than arteriovenous fistulae (AVF). Smoking is already established as a risk factor in lower extremity bypass graft failure used for peripheral vascular disease, but its effect on AVGs remains unclear. We aimed to investigate the relationship of smoking on AVG outcomes. METHODS A 3 year (01/08/2015-01/08/2018) multi-centre retrospective study was carried out on patients receiving an AVG for HD. Data included patient demographics, medical history, operation, type of graft, postoperative course and primary and secondary patency rates. Statistical analyses performed were Kaplan-Meier curves and Cox's proportional hazard regression. RESULTS Fifty-five AVGs were performed (1052 AVF performed) in this period. The most common complication was thrombosis (38.9%). Primary patency at 6, 12 and 24 months were 55%, 45% and 44% respectively. Secondary patency at 6, 12 and 24 months were 63%, 56% and 54% respectively. Smoking was found to be a poor prognostic factor for primary (HR 3.734 (1.818-7.668 95% CI) p < 0.001) and secondary patency (HR 6.238 (2.729-14.257) p < 0.001). Smoking was also significantly associated with graft thrombosis (HR 5.741 (2.380-13.848 95% CI) p < 0.001). DISCUSSION Primary patency rates are lower than previous reports whilst secondary patency is equivalent. Smoking results in a greater risk of thrombosis and poorer primary and secondary patency. This is recognised in vascular surgical grafts, but has not been previously described in AVGs for HD access. Smoking is a modifiable risk factor and as AVGs are typically used for end-stage vascular access patients. Pre-operative strategies to promote smoking cessation, including patient education and prehabilitation should be employed to improve outcomes.
Collapse
Affiliation(s)
- Aaron Kler
- Manchester Centre for Transplantation, Manchester Royal Infirmary, Manchester, UK
| | - Hussein Khambalia
- Manchester Centre for Transplantation, Manchester Royal Infirmary, Manchester, UK
| | - Zulfikar Pondor
- Department of Nephrology, Salford Royal Foundation Trust, Manchester, UK
| | - Rosie Donne
- Department of Nephrology, Salford Royal Foundation Trust, Manchester, UK
| | - Giuseppe Giuffrida
- Manchester Centre for Transplantation, Manchester Royal Infirmary, Manchester, UK
| | - Titus Augustine
- Manchester Centre for Transplantation, Manchester Royal Infirmary, Manchester, UK
| | - Babatunde Campbell
- Manchester Centre for Transplantation, Manchester Royal Infirmary, Manchester, UK
| | - David van Dellen
- Manchester Centre for Transplantation, Manchester Royal Infirmary, Manchester, UK
| |
Collapse
|
4
|
Lee SJ, Lee JS, Kim M, Park SY, Park JH, Park B, Jung WS, Choi JW, Hong JM. Influence of endothelial function and arterial stiffness on the behavior of cervicocephalic arterial dissections: An observational study. Front Neurol 2022; 13:968488. [PMID: 36105775 PMCID: PMC9464973 DOI: 10.3389/fneur.2022.968488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
Background The mechanical and physiological properties of the arterial wall might affect the behavior of spontaneous cervicocephalic arterial dissections (CCAD). We aimed to determine the effects of endothelial function and arterial stiffness on the clinical characteristics and outcomes of CCAD using brachial flow-mediated dilation (FMD) and brachial-ankle pulse wave velocity (PWV). Methods From a single-center database, we identified patients admitted from April 2011 to December 2021 with a diagnosis of CCAD who underwent both FMD and PWV. FMD was classified as normal and decreased according to institutional thresholds. PWV was categorized into tertiles. Comparative and multivariable analyses were performed to determine the effects of FMD and PWV values on major clinical outcomes. Results A total of 146 patients (age: 47 ± 11 years; men: 77.4%) were included. The main presentation was ischemic stroke in 76.7% of the patients, while 23.3% presented with headache or other symptoms. Healing of the dissection was observed in 55.8%. In multivariable analysis, Normal FMD levels (vs. decreased; adjusted OR: 4.52, 95% CI [1.95 −10.52]) were associated with spontaneous healing of the dissection. Highest PWV tertile (vs. lowest; adjusted OR: 17.05, 95% CI [3.07–94.82]) was associated with ischemic presentation. There was a higher ischemic stroke recurrence in the 3rd PWV tertile, and more frequent aneurysmal enlargement in the lowest PWV tertile, but their frequency was low, precluding multivariable analysis. Conclusion In spontaneous CCAD, preserved endothelial function was associated with spontaneous arterial healing. Arterial stiffness is associated with ischemic presentation.
Collapse
Affiliation(s)
- Seong-Joon Lee
- Department of Neurology, Ajou University School of Medicine, Suwon-si, South Korea
- *Correspondence: Seong-Joon Lee
| | - Jin Soo Lee
- Department of Neurology, Ajou University School of Medicine, Suwon-si, South Korea
| | - Min Kim
- Department of Neurology, Ajou University School of Medicine, Suwon-si, South Korea
| | - So Young Park
- Department of Neurology, Ajou University School of Medicine, Suwon-si, South Korea
| | - Ji Hyun Park
- Office of Biostatistics, Medical Research Collaborating Center, Ajou Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon-si, South Korea
| | - Bumhee Park
- Office of Biostatistics, Medical Research Collaborating Center, Ajou Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon-si, South Korea
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon-si, South Korea
| | - Woo Sang Jung
- Department of Radiology, Ajou University School of Medicine, Suwon-si, South Korea
| | - Jin Wook Choi
- Department of Radiology, Ajou University School of Medicine, Suwon-si, South Korea
| | - Ji Man Hong
- Department of Neurology, Ajou University School of Medicine, Suwon-si, South Korea
| |
Collapse
|
5
|
Wang YC, Cai D, Cui XB, Chuang YH, Fay WP, Chen SY. Janus Kinase 3 Deficiency Promotes Vascular Reendothelialization-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:2019-2026. [PMID: 33910370 PMCID: PMC8159884 DOI: 10.1161/atvbaha.121.316293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yung-Chun Wang
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212
| | - Dunpeng Cai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212
- Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Xiao-Bing Cui
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212
| | - Ya-Hui Chuang
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212
| | - William P. Fay
- Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO 65212
- Medicine, University of Missouri School of Medicine, Columbia, MO 65212
- The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65212
| | - Shi-You Chen
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212
- Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO 65212
| |
Collapse
|
6
|
Dubey AK, Kalita J, Chaudhary SK, Misra UK. Impact of anticoagulation status on recanalization and outcome of cerebral venous thrombosis. J Clin Neurosci 2021; 89:43-50. [PMID: 34119293 DOI: 10.1016/j.jocn.2021.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/23/2021] [Accepted: 04/17/2021] [Indexed: 11/26/2022]
Abstract
Effective anticoagulation status may determine the recanalization and outcome of cerebral venous thrombosis (CVT). We report impact of anticoagulation status on recanalization and outcome of CVT. This is a retrospective study on 126 patients with CVT diagnosed on magnetic resonance venography (MRV). Their clinical features and risk factors were noted. The data were retrieved from a prospectively maintained registry, and international normalized ratio (INR) was noted after discharge till 3 months. All the patients were on acenocoumarol. Based on INR value, patients were categorized as Group A (effective anticoagulation INR within the therapeutic range or above) and Group B (ineffective anticoagulation INR > 50% below the therapeutic range). A repeat MRV at 3 months was done for recanalization. Outcome at 3 months was evaluated using modified Rankin Scale (mRS), and categorized as good (mRS ≤ 2) and poor (mRS 2 or more) 101(80.2%) patients were in group A and 25(19.8%) in group B. Their demographic, risk factors, magnetic resonance imaging (MRI) and MRV findings were comparable. On repeat MRV, recanalization occurred in 22/24(91.7%); 15(88%) in group A and 7(100%) in group B. Recanalization was independent of coagulation status. Seven (5.6%) patients died and 107(84.9%) had good outcome; 85(84.2%) in group A and 22(88%) in group B. Kaplan Meier analysis also did not reveal survival or good outcome benefits between the groups. In CVT, outcome and recanalization at 3 months are not dependent on coagulation status. Further prospective studies are needed regarding duration of anticoagulant and its impact on recanalization and outcome.
Collapse
Affiliation(s)
- Ashish K Dubey
- Department of Neurology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Jayantee Kalita
- Department of Neurology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014, India.
| | - Sarvesh K Chaudhary
- Department of Neurology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Usha K Misra
- Apollo Medics Super-specialty Hospitals and Senior consultant neurologist, Vivekanand Polyclinic, and Institute of Medical science, Lucknow 226001, India
| |
Collapse
|
7
|
Remes A, Basha D, Frey N, Wagner A, Müller O. Gene transfer to the vascular system: Novel translational perspectives for vascular diseases. Biochem Pharmacol 2020; 182:114265. [DOI: 10.1016/j.bcp.2020.114265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 01/04/2023]
|
8
|
Wang X, Wang H, He F, Zhang J. In Vitro Cell Migration through Three-Dimensional Interfaces of Varying Depths, Widths, and Curvatures on Micropatterned Polymer Surfaces. ACS APPLIED BIO MATERIALS 2020; 3:7472-7482. [DOI: 10.1021/acsabm.0c00697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xuan Wang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Hefei Innovation Research Institute, Beihang University, 230013 Hefei, China
| | - Hua Wang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Hefei Innovation Research Institute, Beihang University, 230013 Hefei, China
| | - Fang He
- Hefei Innovation Research Institute, Beihang University, 230013 Hefei, China
| | - Jicong Zhang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Hefei Innovation Research Institute, Beihang University, 230013 Hefei, China
- Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, 100083 Beijing, China
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, Beihang University, 100083 Beijing, China
| |
Collapse
|
9
|
Bosseboeuf E, Raimondi C. Signalling, Metabolic Pathways and Iron Homeostasis in Endothelial Cells in Health, Atherosclerosis and Alzheimer's Disease. Cells 2020; 9:cells9092055. [PMID: 32911833 PMCID: PMC7564205 DOI: 10.3390/cells9092055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells drive the formation of new blood vessels in physiological and pathological contexts such as embryonic development, wound healing, cancer and ocular diseases. Once formed, all vessels of the vasculature system present an endothelial monolayer (the endothelium), lining the luminal wall of the vessels, that regulates gas and nutrient exchange between the circulating blood and tissues, contributing to maintaining tissue and vascular homeostasis. To perform their functions, endothelial cells integrate signalling pathways promoted by growth factors, cytokines, extracellular matrix components and signals from mechanosensory complexes sensing the blood flow. New evidence shows that endothelial cells rely on specific metabolic pathways for distinct cellular functions and that the integration of signalling and metabolic pathways regulates endothelial-dependent processes such as angiogenesis and vascular homeostasis. In this review, we provide an overview of endothelial functions and the recent advances in understanding the role of endothelial signalling and metabolism in physiological processes such as angiogenesis and vascular homeostasis and vascular diseases. Also, we focus on the signalling pathways promoted by the transmembrane protein Neuropilin-1 (NRP1) in endothelial cells, its recently discovered role in regulating mitochondrial function and iron homeostasis and the role of mitochondrial dysfunction and iron in atherosclerosis and neurodegenerative diseases.
Collapse
|
10
|
Ren J, Zhou T, Pilli VSS, Phan N, Wang Q, Gupta K, Liu Z, Sheibani N, Liu B. Novel Paracrine Functions of Smooth Muscle Cells in Supporting Endothelial Regeneration Following Arterial Injury. Circ Res 2020; 124:1253-1265. [PMID: 30739581 DOI: 10.1161/circresaha.118.314567] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Regeneration of denuded or injured endothelium is an important component of vascular injury response. Cell-cell communication between endothelial cells and smooth muscle cells (SMCs) plays a critical role not only in vascular homeostasis but also in disease. We have previously demonstrated that PKCδ (protein kinase C-delta) regulates multiple components of vascular injury response including apoptosis of SMCs and production of chemokines, thus is an attractive candidate for a role in SMC-endothelial cells communication. OBJECTIVE To test whether PKCδ-mediated paracrine functions of SMCs influence reendothelialization in rodent models of arterial injury. METHODS AND RESULTS Femoral artery wire injury was performed in SMC-conditional Prkcd knockout mice, and carotid angioplasty was conducted in rats receiving transient Prkcd knockdown or overexpression. SMC-specific knockout of Prkcd impaired reendothelialization, reflected by a smaller Evans blue-excluding area in the knockout compared with the wild-type controls. A similar impediment to reendothelialization was observed in rats with SMC-specific knockdown of Prkcd. In contrast, SMC-specific gene transfer of Prkcd accelerated reendothelialization. In vitro, medium conditioned by AdPKCδ-infected SMCs increased endothelial wound closure without affecting their proliferation. A polymerase chain reaction-based array analysis identified Cxcl1 and Cxcl7 among others as PKCδ-mediated chemokines produced by SMCs. Mechanistically, we postulated that PKCδ regulates Cxcl7 expression through STAT3 (signal transducer and activator of transcription 3) as knockdown of STAT3 abolished Cxcl7 expression. The role of CXCL7 in SMC-endothelial cells communication was demonstrated by blocking CXCL7 or its receptor CXCR2, both significantly inhibited endothelial wound closure. Furthermore, insertion of a Cxcl7 cDNA in the lentiviral vector that carries a Prkcd shRNA overcame the adverse effects of Prkcd knockdown on reendothelialization. CONCLUSIONS SMCs promote reendothelialization in a PKCδ-dependent paracrine mechanism, likely through CXCL7-mediated recruitment of endothelial cells from uninjured endothelium.
Collapse
Affiliation(s)
- Jun Ren
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Ting Zhou
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Vijaya Satish Sekhar Pilli
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Noel Phan
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Qiwei Wang
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Kartik Gupta
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| | - Zhenjie Liu
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.).,Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University (Z.L.)
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison (N.S.)
| | - Bo Liu
- From the Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison (J.R., T.Z., V.S.S.P., N.P., Q.W., K.G., Z.L., B.L.)
| |
Collapse
|
11
|
Ferrone M, Cheng Y, Granada JF. Current concepts regarding drug dosing for peripheral stents. THE JOURNAL OF CARDIOVASCULAR SURGERY 2019; 60:439-449. [PMID: 31062571 DOI: 10.23736/s0021-9509.19.10995-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Drug-eluting stent (DES) are the mainstay therapy for the treatment of coronary artery disease. Stent design and drug-elution strategies have evolved over the years leading to the last generation DES which shows optimal safety and efficacy outcome. Peripheral arteries have different mechanical and biological features and the lessons learned from the coronary field have been difficult to introduce into the development of peripheral vascular technologies. First, due to its complex biomechanical behavior the use of metallic stents is limited in some vascular segments (i.e., distal superficial fermoral artery [SFA]). Also, peripheral vascular atherosclerosis is different containing higher levels of plaque burden and calcium. Finally, peripheral arterial disease tends to be more aggressive including longer lesions and higher incidence of total chronic occlusion. In general terms, restenosis in the peripheral vascular territory is more aggressive and occurs at a later time (~12 months) requiring a different pharmacokinetic profile compared to coronary technologies. Several strategies have been evaluated in the peripheral arteries raging from the bare metal stent to the drug coated balloon and drug eluting stent with outcome varying depending on the different field of application (i.e. SFA and below-the-knee). Results coming from the clinical trial are encouraging but further studies and direct comparison among the different technologies are demanded to determine the best therapy for peripheral vascular disease.
Collapse
Affiliation(s)
- Marco Ferrone
- Cardiovascular Research Foundation, Skirball Center for Innovation, Orangeburg, NY, USA.,Federico II University of Naples, Naples, Italy
| | - Yanping Cheng
- Cardiovascular Research Foundation, Skirball Center for Innovation, Orangeburg, NY, USA
| | - Juan F Granada
- Cardiovascular Research Foundation, Skirball Center for Innovation, Orangeburg, NY, USA -
| |
Collapse
|
12
|
Spangler TA, Katzman SA. Pathological Safety Assessment in Preclinical Neurothrombectomy Studies. Toxicol Pathol 2019; 47:264-279. [PMID: 30832552 DOI: 10.1177/0192623319826066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The design, production, and preclinical testing of neurothrombectomy devices is in a burgeoning phase as the demand escalates for safe and reliable treatment options following neurovascular stroke. Currently, there is a paucity of published data describing the development of iatrogenic vascular lesions occurring secondary to neurothrombectomy procedures. In an effort to test new devices, demonstrate device safety, satisfy regulatory requirements, and develop an understanding of the potential for associated vascular pathology, investigators are establishing appropriate methodology in suitable animal models. Significant challenges exist in identifying a single animal species that can be consistently utilized in all phases of device development. These aforementioned challenges are underscored by the intricacies of neurovascular pathology, thrombovascular interactions, and vascular responses to injury.
Collapse
Affiliation(s)
| | - Scott A Katzman
- 2 William R. Pritchard Veterinary Medical Teaching Hospital, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
13
|
Peschillo S, Tomasello A, Diana F, Hernandez D, Toccaceli G, Rosal-Fontana M, Esteves Coelho M, Missori P. Comparison of Subacute Vascular Damage Caused by ADAPT versus Stent Retriever Devices after Thrombectomy in Acute Ischemic Stroke: Histological and Ultrastructural Study in an Animal Model. INTERVENTIONAL NEUROLOGY 2018; 7:501-512. [PMID: 30410530 DOI: 10.1159/000491028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022]
Abstract
Objectives To assess the delayed (15 days) histological and ultrastructural changes occurring following endovascular treatment with a direct aspiration first pass technique (ADAPT) or stent retrievers (SRs) and to compare the findings in order to determine which is the least harmful technique and what changes occur. Materials and Methods Damage to the wall of swine extracranial arteries was evaluated after ADAPT with the Penumbra system or thrombectomy with various SRs. The procedures were performed using two pigs as animal models; extracranial cervical arteries were selected based on their diameters in order to reproduce the procedures as in human intracranial arteries, and endovascular thrombectomies were done after the injection of autologous thrombi. Two weeks later, the animals were euthanized, and 60 arterial samples were obtained for analysis by optical and electron microscopy. Results Optical and electron microscopy revealed that both techniques cause, in different way, alterations to the structure of the vessel wall. Conclusions Both techniques caused damage to the vessel wall. The main damages were localized at the level of the tunica media and adventitia, instead of the tunica intima as in the acute phase. Further investigation is required to better understand whether these alterations could have chronic consequences.
Collapse
Affiliation(s)
| | | | - Francesco Diana
- Interventional Neuroradiology, Policlinico Umberto I, Rome, Italy
| | - David Hernandez
- Interventional Neuroradiology, Vall d'Hebron Hospital, Barcelona, Spain
| | - Giada Toccaceli
- Endovascular Neurosurgery, Policlinico Umberto I, Rome, Italy
| | | | | | | |
Collapse
|
14
|
Li J, Zhang K, Huang N. Engineering Cardiovascular Implant Surfaces to Create a Vascular Endothelial Growth Microenvironment. Biotechnol J 2017; 12. [PMID: 28941232 DOI: 10.1002/biot.201600401] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 09/14/2017] [Indexed: 12/16/2022]
Abstract
Cardiovascular disease (CVD) is generally accepted as the leading cause of morbidity and mortality worldwide, and an increasing number of patients suffer from atherosclerosis and thrombosis annually. To treat these disorders and prolong the sufferers' life, several cardiovascular implants have been developed and applied clinically. Nevertheless, thrombosis and hyperplasia at the site of cardiovascular implants are recognized as long-term problems in the practice of interventional cardiology. Here, we start this review from the clinical requirement of the implants, such as anti-hyperplasia, anti-thrombosis, and pro-endothelialization, wherein particularly focus on the natural factors which influence functional endothelialization in situ, including the healthy smooth muscle cells (SMCs) environment, blood flow shear stress (BFSS), and the extracellular matrix (ECM) microenvironment. Then, the currently available strategies on surface modification of cardiovascular biomaterials to create vascular endothelial growth microenvironment are introduced as the main topic, e.g., BFSS effect simulation by surface micro-patterning, ECM rational construction and SMCs phenotype maintain. Finally, the prospects for extending use of the in situ construction of endothelial cells growth microenvironment are discussed and summarized in designing the next generation of vascular implants.
Collapse
Affiliation(s)
- Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China.,Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Kun Zhang
- Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China.,School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Nan Huang
- Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| |
Collapse
|
15
|
Neue Mechanismen zur Verhinderung der myointimalen Hyperplasie. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2017. [DOI: 10.1007/s00398-017-0153-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Nuhn H, Blanco CE, Desai TA. Nanoengineered Stent Surface to Reduce In-Stent Restenosis in Vivo. ACS APPLIED MATERIALS & INTERFACES 2017; 9:19677-19686. [PMID: 28574242 DOI: 10.1021/acsami.7b04626] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In-stent restenosis (ISR) is the leading cause of stent failure and is a direct result of a dysfunctional vascular endothelium and subsequent overgrowth of vascular smooth muscle tissue. TiO2 nanotubular (NT) arrays have been shown to affect vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) in vitro by accelerating VEC cell proliferation and migration while suppressing VSMCs. This study investigates for the first time the potentially beneficial effects of TiO2 NT arrays on vascular tissue in vivo. TiO2 NT arrays (NT diameter: 90 ± 5 nm, height: 1800 ± 300 nm) were grown on the surface of titanium stents and characterized in terms of surface morphology and stability. Stents were implanted into the iliofemoral artery using an overinflation model (rabbit). After 28 days, stenosis rates were determined. The data show a statistically significant reduction of stenosis by 30% compared to the control. Tissue in the presence of TiO2 NTs appears more mature, and less neointima is present between struts. In addition, the extra cellular matrix secreted by cells at the interface of the NT arrays shows complete integration into the nanostructured surface. These results document the accelerated restoration of a functional endothelium in the presence of TiO2 NT arrays and substantiate their beneficial impact on vascular tissue in vivo. Our findings suggest that TiO2 NT arrays can be used as a drug-free approach for keeping stents patent long-term and have the potential to address ISR.
Collapse
Affiliation(s)
- Harald Nuhn
- The Alfred E. Mann Institute for Biomedical Engineering at the University of Southern California , 1042 Downey Way, DRB Building, Suite 101, Los Angeles, California 90089-1112, United States
| | - Cesar E Blanco
- The Alfred E. Mann Institute for Biomedical Engineering at the University of Southern California , 1042 Downey Way, DRB Building, Suite 101, Los Angeles, California 90089-1112, United States
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences and The UC Berkeley-UCSF Graduate Group in Bioengineering, University of California-San Francisco , San Francisco, California 94158, United States
| |
Collapse
|
17
|
Luo W, Guth CM, Jolayemi O, Duvall CL, Brophy CM, Cheung-Flynn J. Subfailure Overstretch Injury Leads to Reversible Functional Impairment and Purinergic P2X7 Receptor Activation in Intact Vascular Tissue. Front Bioeng Biotechnol 2016; 4:75. [PMID: 27747211 PMCID: PMC5040722 DOI: 10.3389/fbioe.2016.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/13/2016] [Indexed: 11/30/2022] Open
Abstract
Vascular stretch injury is associated with blunt trauma, vascular surgical procedures, and harvest of human saphenous vein for use in vascular bypass grafting. A model of subfailure overstretch in rat abdominal aorta was developed to characterize surgical vascular stretch injury. Longitudinal stretch of rat aorta was characterized ex vivo. Stretch to the haptic endpoint, where the tissues would no longer lengthen, occurred at twice the resting length. The stress produced at this length was greater than physiologic mechanical forces but well below the level of mechanical disruption. Functional responses were determined in a muscle bath, and this subfailure overstretch injury led to impaired smooth muscle function that was partially reversed by treatment with purinergic receptor (P2X7R) antagonists. These data suggest that vasomotor dysfunction caused by subfailure overstretch injury may be due to the activation of P2X7R. These studies have implications for our understanding of mechanical stretch injury of blood vessels and offer novel therapeutic opportunities.
Collapse
Affiliation(s)
- Weifeng Luo
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Christy M. Guth
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Olukemi Jolayemi
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Colleen Marie Brophy
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | |
Collapse
|
18
|
Derkacz A, Protasiewicz M, Rola P, Podgorska K, Szymczyszyn A, Gutherc R, Poręba R, Doroszko A. Effects of intravascular low-level laser therapy during coronary intervention on selected growth factors levels. Photomed Laser Surg 2016; 32:582-7. [PMID: 25302462 DOI: 10.1089/pho.2013.3700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE The objective of this study was to evaluate the effect of intravascular low-level laser therapy (LLLT) on selected growth factor levels in subjects undergoing percutaneous coronary interventions (PCI). BACKGROUND DATA Restenosis remains the main problem with the long-term efficacy of PCI, and growth factors are postulated to play a crucial role in the restenosis cascade. MATERIALS AND METHODS In a randomized prospective study, an 808 nm LLLT (100 mW/cm2, continuous wave laser, 9 J/cm2, illuminated area 1.6-2.5 cm2) was delivered intracoronarily to patients during PCI. Fifty-two patients underwent irradiation with laser light, and 49 constituted the control group. In all individuals, serum levels of insulin-like growth factor-1 (IGF-1), vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1), and fibroblast growth factor-2 (FGF-2) were measured before angioplasty, then 6 and 12 h and 1 month after the procedure. In all patients, a control angiography was performed 6 months later. RESULTS There were no significant differences in IGF-1 and VEGF levels between the groups. While evaluating FGF-2, we observed its significantly lower levels in the irradiated patients during each examination. There was a significant increase in TGF-β1 level in control group after 12 h of observation. In the irradiated individuals, control angiography revealed smaller late lumen loss and smaller late lumen loss index as compared with the control group. The restenosis rate was 15.0% in the treated group, and 32.4% in the control group, respectively. CONCLUSIONS LLLT decreases levels of TGF-β1 and FGF-2 in patients undergoing coronary intervention, which may explain smaller neointima formation.
Collapse
Affiliation(s)
- Arkadiusz Derkacz
- 1 Department of Internal Medicine and Hypertension, Wroclaw Medical University , Wroclaw, Poland
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Adamo RF, Fishbein I, Zhang K, Wen J, Levy RJ, Alferiev IS, Chorny M. Magnetically enhanced cell delivery for accelerating recovery of the endothelium in injured arteries. J Control Release 2015; 222:169-75. [PMID: 26704936 DOI: 10.1016/j.jconrel.2015.12.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 01/27/2023]
Abstract
Arterial injury and disruption of the endothelial layer are an inevitable consequence of interventional procedures used for treating obstructive vascular disease. The slow and often incomplete endothelium regrowth after injury is the primary cause of serious short- and long-term complications, including thrombosis, restenosis and neoatherosclerosis. Rapid endothelium restoration has the potential to prevent these sequelae, providing a rationale for developing strategies aimed at accelerating the reendothelialization process. The present studies focused on magnetically guided delivery of endothelial cells (EC) functionalized with biodegradable magnetic nanoparticles (MNP) as an experimental approach for achieving rapid and stable cell homing and expansion in stented arteries. EC laden with polylactide-based MNP exhibited strong magnetic responsiveness, capacity for cryopreservation and rapid expansion, and the ability to disintegrate internalized MNP in both proliferating and contact-inhibited states. Intracellular decomposition of BODIPY558/568-labeled MNP monitored non-invasively based on assembly state-dependent changes in the emission spectrum demonstrated cell proliferation rate-dependent kinetics (average disassembly rates: 6.6±0.8% and 3.6±0.4% per day in dividing and contact-inhibited EC, respectively). With magnetic guidance using a transient exposure to a uniform 1-kOe field, stable localization and subsequent propagation of MNP-functionalized EC, markedly enhanced in comparison to non-magnetic delivery conditions, were observed in stented rat carotid arteries. In conclusion, magnetically guided delivery is a promising experimental strategy for accelerating endothelial cell repopulation of stented blood vessels after angioplasty.
Collapse
Affiliation(s)
- Richard F Adamo
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ilia Fishbein
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kehan Zhang
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Justin Wen
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Robert J Levy
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ivan S Alferiev
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Michael Chorny
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Brenner C, Kränkel N, Kühlenthal S, Israel L, Remm F, Fischer C, Herbach N, Speer T, Grabmaier U, Laskowski A, Gross L, Theiss H, Wanke R, Landmesser U, Franz WM. Short-term inhibition of DPP-4 enhances endothelial regeneration after acute arterial injury via enhanced recruitment of circulating progenitor cells. Int J Cardiol 2014; 177:266-75. [PMID: 25499391 DOI: 10.1016/j.ijcard.2014.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/06/2014] [Accepted: 09/15/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Endothelial injuries regularly occur in atherosclerosis and during interventional therapies of the arterial occlusive disease. Disturbances in the endothelial integrity can lead to insufficient blood supply and bear the risk of thrombus formation and acute vascular occlusion. At present, effective therapeutics to restore endothelial integrity are barely available. We analyzed the effect of pharmacological DPP-4-inhibition by Sitagliptin on endogenous progenitor cell-based endothelial regeneration via the SDF-1α/CXCR4-axis after acute endothelial damage in a mouse model of carotid injury. METHODS AND RESULTS Induction of a defined endothelial injury was performed in the carotid artery of C57Bl/6 mice which led to a local upregulation of SDF-1α expression. Animals were treated with placebo, Sitagliptin or Sitagliptin+AMD3100. Using mass spectrometry we could prove that Sitagliptin prevented cleavage of the chemokine SDF-1α. Accordingly, increased SDF-1α concentrations enhanced recruitment of systemically applied and endogenous circulating CXCR4+ progenitor cells to the site of vascular injury followed by a significantly accelerated reendothelialization as compared to placebo-treated animals. Improved endothelial recovery, as well as recruitment of circulating CXCR4+ progenitor cells (CD133+, Flk1+), was reversed by CXCR4-antagonization through AMD3100. In addition, short-term Sitagliptin treatment did not significantly promote neointimal or medial hyperplasia. CONCLUSION Sitagliptin can accelerate endothelial regeneration after acute endothelial injury. DPP-4 inhibitors prevent degradation of the chemokine SDF-1α and thus improve the recruitment of regenerative circulating CXCR4+ progenitor cells which mediate local endothelial cell proliferation without adversely affecting vessel wall architecture.
Collapse
Affiliation(s)
- Christoph Brenner
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland; Department of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| | - Nicolle Kränkel
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland
| | - Sarah Kühlenthal
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Lars Israel
- Institute of Molecular Biology, Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| | - Friederike Remm
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Cornelia Fischer
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Nadja Herbach
- Institute of Veterinary Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Timo Speer
- Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland; Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Ulrich Grabmaier
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Alexandra Laskowski
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Lisa Gross
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Hans Theiss
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Ulf Landmesser
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, Cardiovascular Research, University of Zurich, Campus Irchel, Zurich, Switzerland
| | - Wolfgang-Michael Franz
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany; Department of Internal Medicine III, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
21
|
Yin K, Agrawal DK. High-density lipoprotein: a novel target for antirestenosis therapy. Clin Transl Sci 2014; 7:500-11. [PMID: 25043950 DOI: 10.1111/cts.12186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Restenosis is an integral pathological process central to the recurrent vessel narrowing after interventional procedures. Although the mechanisms for restenosis are diverse in different pathological conditions, endothelial dysfunction, inflammation, vascular smooth muscle cell (SMC) proliferation, and myofibroblasts transition have been thought to play crucial role in the development of restenosis. Indeed, there is an inverse relationship between high-density lipoprotein (HDL) levels and risk for coronary heart disease (CHD). However, relatively studies on the direct assessment of HDL effect on restenosis are limited. In addition to involvement in the cholesterol reverse transport, many vascular protective effects of HDL, including protection of endothelium, antiinflammation, antithrombus actions, inhibition of SMC proliferation, and regulation by adventitial effects may contribute to the inhibition of restenosis, though the exact relationships between HDL and restenosis remain to be elucidated. This review summarizes the vascular protective effects of HDL, emphasizing the potential role of HDL in intimal hyperplasia and vascular remodeling, which may provide novel prophylactic and therapeutic strategies for antirestenosis.
Collapse
Affiliation(s)
- Kai Yin
- Center for Clinical & Translational Science, Creighton University School of Medicine, Omaha, Nebraska, USA
| | | |
Collapse
|
22
|
Cubbon RM, Mercer BN, Sengupta A, Kearney MT. Importance of insulin resistance to vascular repair and regeneration. Free Radic Biol Med 2013; 60:246-63. [PMID: 23466555 DOI: 10.1016/j.freeradbiomed.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/22/2013] [Accepted: 02/23/2013] [Indexed: 01/14/2023]
Abstract
Metabolic insulin resistance is apparent across a spectrum of clinical disorders, including obesity and diabetes, and is characterized by an adverse clustering of cardiovascular risk factors related to abnormal cellular responses to insulin. These disorders are becoming increasingly prevalent and represent a major global public health concern because of their association with significant increases in atherosclerosis-related mortality. Endogenous repair mechanisms are thought to retard the development of vascular disease, and a growing evidence base supports the adverse impact of the insulin-resistant phenotype upon indices of vascular repair. Beyond the impact of systemic metabolic changes, emerging data from murine studies also provide support for abnormal insulin signaling at the level of vascular cells in retarding vascular repair. Interrelated pathophysiological factors, including reduced nitric oxide bioavailability, oxidative stress, altered growth factor activity, and abnormal intracellular signaling, are likely to act in conjunction to impede vascular repair while also driving vascular damage. Understanding of these processes is shaping novel therapeutic paradigms that aim to promote vascular repair and regeneration, either by recruiting endogenous mechanisms or by the administration of cell-based therapies.
Collapse
Affiliation(s)
- Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre, LIGHT Laboratories, The University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | |
Collapse
|
23
|
Albiero M, Avogaro A, Fadini GP. Restoring stem cell mobilization to promote vascular repair in diabetes. Vascul Pharmacol 2013; 58:253-8. [PMID: 23369723 DOI: 10.1016/j.vph.2013.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/12/2013] [Accepted: 01/15/2013] [Indexed: 12/30/2022]
Abstract
Diabetes triggers endothelial dysfunction, which is linked to increased risk of cardiovascular diseases. Stem and progenitor cells from the bone marrow are involved in the maintenance of vascular integrity. Diabetic patients show a dysfunction of these cells, which might represent a novel pathophysiological mechanism of vascular disease. Specifically, stem and progenitor cells fail to egress from the bone marrow (BM) due to BM pathological alterations and unresponsiveness to mobilizing stimuli. In this review, we describe impaired stem cell mobilization in diabetes as a mechanism of failed vascular repair and we provide evidence that pharmacological strategies can restore mobilization. We discuss recent advances in the knowledge of aberrant organization of the diabetic BM and its implications for impaired mobilization. Finally, we describe in detail the pharmacological exploitation of the G-CSF/DPP-4(CD26)/SDF-1α axis as a novel strategy to improve mobilization and attain vascular repair in diabetes.
Collapse
Affiliation(s)
- Mattia Albiero
- Venetian Institute of Molecular Medicine, Laboratory of Experimental Diabetology, 35100 Padova, Italy
| | | | | |
Collapse
|
24
|
Gucu A, Cavusoglu I, Bozkurt O, Eris C, Toktas F, Goncu T, Ozyazicioglu A. Effects of temporary vascular occluder poloxamer 407 gel on the endothelium. J Cardiothorac Surg 2013; 8:16. [PMID: 23339359 PMCID: PMC3577636 DOI: 10.1186/1749-8090-8-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 01/03/2013] [Indexed: 12/02/2022] Open
Abstract
Background Coronary occlusion techniques during OPCAB may lead to an endothelial damage to the target vessel. The adverse effects of these techniques are well-known, and researches have been trying to find out new materials to occlude the coronary artery without an endothelial damage. In the present study, we investigate to the endothelial damage in the rat aorta which is occluded by Poloxamer 407 gel. Methods Forty-five rats were randomized in three groups: (1) segment of the aorta was occluded with Poloxamer 407 gel in P 407 group; (2) segment of the aorta was occluded with microvascular clamp in MV clamp group; and (3) no onclusion was available in the Control group. The rats were sacrificed of observation, and a 15mm segment of the aorta was obtained as a specimen. Integrity of the endothelial lining was observed with a scanning electron microscopy. Results Scanning electron microscopy revealed a statistically significant difference among the 3 groups (p<0,001) using the SPSS 13.0 test. No difference was found between the Control group and the P 407 group (p=0,059). The differences between MV clamp–Control group (p<0,001) and MV clamp–P 407 group were statistically significant (p<0,002). Conclusions We suggest that Poloxamer 407 gel occlusion may be a safer and more effective method compared to the microvascular clamp occlusion.
Collapse
Affiliation(s)
- Arif Gucu
- Department of Cardiovascular Surgery, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey.
| | | | | | | | | | | | | |
Collapse
|
25
|
Berra-Romani R, Raqeeb A, Torres-Jácome J, Guzman-Silva A, Guerra G, Tanzi F, Moccia F. The mechanism of injury-induced intracellular calcium concentration oscillations in the endothelium of excised rat aorta. J Vasc Res 2011; 49:65-76. [PMID: 21997119 DOI: 10.1159/000329618] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 05/20/2011] [Indexed: 01/13/2023] Open
Abstract
Endothelial injury is the primary event that leads to a variety of severe vascular disorders. Mechanical injury elicits a Ca(2+) response in the endothelium of excised rat aorta, which comprises an initial Ca(2+) release from inositol-1,4,5-trisphosphate (InsP(3))-sensitive stores followed by a long-lasting decay phase due to Ca(2+) entry through uncoupled connexons. The Ca(2+) signal may also adopt an oscillatory pattern, the molecular underpinnings of which are unclear. In the light of the role played by Ca(2+) spiking in tissue regeneration, this study aimed to unveil the mechanisms underlying injury-induced Ca(2+) oscillations. The latter reversibly ceased upon removal of extracellular Ca(2+) or addition of the gap junction blockers heptanol, 18 α,β-glycyrrhetinic acid, La(3+) and Ni(2+), but were insensitive to BTP-2 and SKF 96365. The spiking response was abolished by inhibiting the Ca(2+) entry mode of the Na(+)/Ca(2+) exchanger (NCX). The InsP(3)-producing agonist ATP resumed Ca(2+) oscillations in silent cells, while the phospholipase C inhibitor U73122 suppressed them. Injury-induced Ca(2+) transients were prevented by the sarcoplasmic-endoplasmic reticulum calcium ATPase (SERCA) blockers thapsigargin and cyclopiazonic acid, while they were unaffected by suramin and genistein. These data show for the first time that the coordinated interplay between NCX-mediated Ca(2+) entry and InsP(3)-dependent Ca(2+) release contributes to injury-induced intracellular Ca(2+) concentration oscillations.
Collapse
Affiliation(s)
- Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, México.
| | | | | | | | | | | | | |
Collapse
|
26
|
Kamat N, Nguyen-Ehrenreich KLT, Hsu SH, Ma AP, Sinn I, Coleman L, Tai J. Characterization of Vascular Injury Responses to Stent Insertion in an Ex-vivo Arterial Perfusion Model. J Vasc Interv Radiol 2011; 22:193-202. [DOI: 10.1016/j.jvir.2010.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 05/30/2010] [Accepted: 10/12/2010] [Indexed: 11/28/2022] Open
|
27
|
Kim YM, Pae HO, Park JE, Lee YC, Woo JM, Kim NH, Choi YK, Lee BS, Kim SR, Chung HT. Heme oxygenase in the regulation of vascular biology: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2011; 14:137-67. [PMID: 20624029 PMCID: PMC2988629 DOI: 10.1089/ars.2010.3153] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heme oxygenases (HOs) are the rate-limiting enzymes in the catabolism of heme into biliverdin, free iron, and carbon monoxide. Two genetically distinct isoforms of HO have been characterized: an inducible form, HO-1, and a constitutively expressed form, HO-2. HO-1 is a kind of stress protein, and thus regarded as a sensitive and reliable indicator of cellular oxidative stress. The HO system acts as potent antioxidants, protects endothelial cells from apoptosis, is involved in regulating vascular tone, attenuates inflammatory response in the vessel wall, and participates in angiogenesis and vasculogenesis. Endothelial integrity and activity are thought to occupy the central position in the pathogenesis of cardiovascular diseases. Cardiovascular disease risk conditions converge in the contribution to oxidative stress. The oxidative stress leads to endothelial and vascular smooth muscle cell dysfunction with increases in vessel tone, cell growth, and gene expression that create a pro-thrombotic/pro-inflammatory environment. Subsequent formation, progression, and obstruction of atherosclerotic plaque may result in myocardial infarction, stroke, and cardiovascular death. This background provides the rationale for exploring the potential therapeutic role for HO system in the amelioration of vascular inflammation and prevention of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Young-Myeong Kim
- Vascular System Research Center and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Kangwon-do, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lenfant F, Trémollières F, Gourdy P, Arnal JF. Timing of the vascular actions of estrogens in experimental and human studies: why protective early, and not when delayed? Maturitas 2010; 68:165-73. [PMID: 21167666 DOI: 10.1016/j.maturitas.2010.11.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 11/18/2010] [Accepted: 11/18/2010] [Indexed: 01/27/2023]
Abstract
Estrogens, and in particular 17β-estradiol (E2), play a pivotal role in sexual development and reproduction and are also implicated in a large number of physiological processes including the cardiovascular system. Although epidemiological studies and Nurses' Health Study suggested, and all animal models of early atheroma clearly demonstrated a vasculoprotective action of both endogenous and exogenous estrogens, the Women's Health Initiative did not confirm the preventive action of estrogens against coronary heart disease (CHD). However, women who initiated hormone therapy closer to menopause tended to have reduced CHD risk compared with increased CHD risk among women more distant from menopause. Thus, it is now mandatory to try to understand the mechanisms that could have influenced the actions of estrogens at various stages of atherosclerosis and/or of life. In this current review, we will summarize our understanding of the potential cellular targets and mechanisms of the vasculoprotective actions of estrogens, as well as of the lack of action of estrogens when administered after a period of hormonal deprivation. The mechanisms of the aggravating role of progestogens such as medroxyprogesterone acetate will be considered. Finally, we will analyze the possibilities to uncouple some beneficial from other undesirable actions following the partial/selective activation of estrogen receptors.
Collapse
Affiliation(s)
- Françoise Lenfant
- INSERM U1048-I2MC, Faculté de Médecine, Université de Toulouse III et CHU de Toulouse, Toulouse, France.
| | | | | | | |
Collapse
|
29
|
Kim YM, Pae HO, Park JE, Lee YC, Woo JM, Kim NH, Choi YK, Lee BS, Kim SR, Chung HT. Heme oxygenase in the regulation of vascular biology: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2010. [PMID: 20624029 DOI: 10.1089/ars.2010.31532988629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heme oxygenases (HOs) are the rate-limiting enzymes in the catabolism of heme into biliverdin, free iron, and carbon monoxide. Two genetically distinct isoforms of HO have been characterized: an inducible form, HO-1, and a constitutively expressed form, HO-2. HO-1 is a kind of stress protein, and thus regarded as a sensitive and reliable indicator of cellular oxidative stress. The HO system acts as potent antioxidants, protects endothelial cells from apoptosis, is involved in regulating vascular tone, attenuates inflammatory response in the vessel wall, and participates in angiogenesis and vasculogenesis. Endothelial integrity and activity are thought to occupy the central position in the pathogenesis of cardiovascular diseases. Cardiovascular disease risk conditions converge in the contribution to oxidative stress. The oxidative stress leads to endothelial and vascular smooth muscle cell dysfunction with increases in vessel tone, cell growth, and gene expression that create a pro-thrombotic/pro-inflammatory environment. Subsequent formation, progression, and obstruction of atherosclerotic plaque may result in myocardial infarction, stroke, and cardiovascular death. This background provides the rationale for exploring the potential therapeutic role for HO system in the amelioration of vascular inflammation and prevention of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Young-Myeong Kim
- Vascular System Research Center and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Kangwon-do, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Arnal JF, Fontaine C, Billon-Galés A, Favre J, Laurell H, Lenfant F, Gourdy P. Estrogen Receptors and Endothelium. Arterioscler Thromb Vasc Biol 2010; 30:1506-12. [DOI: 10.1161/atvbaha.109.191221] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogens, and in particular 17β-estradiol (E2), play a pivotal role in sexual development and reproduction and are also implicated in a large number of physiological processes, including the cardiovascular system. Both acetylcholine-induced and flow-dependent vasodilation are preserved or potentiated by estrogen treatment in both animal models and humans. Indeed, E2 increases the endothelial production of nitric oxide and prostacyclin and prevents early atheroma through endothelial-mediated mechanisms. Furthermore, whereas it prevents endothelial activation, E2 potentiates the ability of several subpopulations of the circulating or resident immune cells to produce proinflammatory cytokines. The balance between these 2 actions could determine the final effect in a given pathophysiological process. E2 also promotes endothelial healing, as well as angiogenesis. Estrogen actions are essentially mediated by 2 molecular targets: estrogen receptor-α (ERα) and ERβ. The analysis of mouse models targeted for ERα or ERβ demonstrated a prominent role of ERα in vascular biology. ERα directly modulates transcription of target genes through 2 activation functions (AFs), AF-1 and AF-2. Interestingly, an AF-1-deficient ERα isoform can be physiologically expressed in the endothelium and appears sufficient to mediate most of the vasculoprotective actions of E2. In contrast, AF-1 is necessary for the E2 actions in reproductive targets. Thus, it appears conceivable to uncouple the vasculoprotective and sexual actions with appropriate selective ER modulators.
Collapse
Affiliation(s)
- Jean-François Arnal
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| | - Audrey Billon-Galés
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| | - Julie Favre
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| | - Henrik Laurell
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- From INSERM U858-I2MC, Faculté de Médecine, Université de Toulouse et CHU de Toulouse, Toulouse, France
| |
Collapse
|
31
|
Hui S, Brunt KR, Husain M. Temporal and spatial regulation of histone deacetylase-7 and beta-catenin in endothelial cells. Circ Res 2010; 106:1180-3. [PMID: 20395600 DOI: 10.1161/circresaha.110.219345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
32
|
Toutain CE, Filipe C, Billon A, Fontaine C, Brouchet L, Guéry JC, Gourdy P, Arnal JF, Lenfant F. Estrogen receptor alpha expression in both endothelium and hematopoietic cells is required for the accelerative effect of estradiol on reendothelialization. Arterioscler Thromb Vasc Biol 2009; 29:1543-50. [PMID: 19608969 DOI: 10.1161/atvbaha.109.192849] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE E2 accelerates reendothelialization through estrogen receptor alpha (ER alpha), and we now aimed at defining the precise local and systemic cellular actors of this process. METHODS AND RESULTS The respective roles of endothelial and hematopoietic targets of E2 were investigated in a mouse carotid injury model, using confocal microscopy, to follow endothelium repair. Grafting ER alpha(-/-) mice with ER alpha(+/+) bone marrow (BM) was not sufficient to restore the accelerative effect of E2 on reendothelialization, demonstrating the necessary role of extrahematopoietic ER alpha. Using an endothelial-specific inactivation of ER alpha (Cre-Lox system), we showed that endothelial ER alpha plays a pivotal role in this E2 action. Conversely, in ER alpha(+/+) grafted with ER alpha(-/-) BM, the E2 regenerative effect was abolished, demonstrating that ER alpha-expressing hematopoietic cells are also needed. As eNOS expression in BM was required for this action, both endothelial progenitor cells and platelets could be the hematopoietic targets that participate to this beneficial E2 effect. CONCLUSIONS We demonstrate that endothelial ER alpha plays a pivotal role in E2-mediated reendothelialization. However, endothelial targeting alone is not sufficient because the concomitant stimulation of a subpopulation of BM ER alpha is necessary. This cooperation should be taken into account in strategies aimed at optimizing in-stent reendothelialization.
Collapse
Affiliation(s)
- Céline E Toutain
- Institut National de la Santé et de la Recherche Médicale, U858, IFR150, and Université de Toulouse III, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Desouza CV, Gerety M, Hamel FG. Effects of a PPAR-gamma agonist, on growth factor and insulin stimulated endothelial cells. Vascul Pharmacol 2009; 51:162-8. [PMID: 19520186 DOI: 10.1016/j.vph.2009.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 04/08/2009] [Accepted: 05/28/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE PPAR-gamma agonists such as thiazolidinediones, used in patients with insulin resistance have been shown to reduce neointimal hyperplasia in the short term. However recent studies suggest increased cardiovascular risk for some thiazolidinediones. Longer-term animal studies show inhibition of endothelial regrowth post endothelial injury which may account for some of the increased risk. We studied the effect of pioglitazone on VEGF, FGF and insulin stimulated endothelial cells to determine if this was a mechanism of inhibition of endothelial regrowth. METHODS AND RESULTS FGF/VEGF stimulated human umbilical vein endothelial cell (HUVEC) proliferation and apoptosis was measured, in vitro, in the presence and absence of hyperinsulinemia, with and without treatment with the PPAR-gamma agonist pioglitazone. Activation of ERK 1/2 and p38MAPK was measured under the same conditions. There was 40% decrease in proliferation with pioglitazone in VEGF stimulated cells, which was reversed by insulin. ERK 1/2 activation was decreased by pioglitazone in VEGF stimulated cells and was partially reversed by insulin. p38MAPK activation was increased by pioglitazone and was unaffected by insulin or VEGF. Pioglitazone also increased endothelial cell apoptosis. CONCLUSION PPAR-gamma agonists may have detrimental cardiovascular effects post angioplasty especially in patients with insulin resistance. We have shown that one of the mechanisms may be inhibition of endothelial regrowth and re-endothelialization by inhibition of VEGF/FGF stimulation of the ERK 1/2 pathways in endothelial cells.
Collapse
Affiliation(s)
- Cyrus V Desouza
- Omaha Veterans Affairs Medical Center, United States; University of Nebraska Medical Center, United States.
| | | | | |
Collapse
|
34
|
Punchard MA, O'Cearbhaill ED, Mackle JN, McHugh PE, Smith TJ, Stenson-Cox C, Barron V. Evaluation of Human Endothelial Cells Post Stent Deployment in a Cardiovascular Simulator In Vitro. Ann Biomed Eng 2009; 37:1322-30. [PMID: 19408120 DOI: 10.1007/s10439-009-9701-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 04/10/2009] [Indexed: 12/28/2022]
Affiliation(s)
- Marie A Punchard
- National Centre for Biomedical Engineering, Orbsen Building, National University of Ireland, Galway, University Road, Galway, Ireland
| | | | | | | | | | | | | |
Collapse
|
35
|
Filipe C, Lam Shang Leen L, Brouchet L, Billon A, Benouaich V, Fontaine V, Gourdy P, Lenfant F, Arnal JF, Gadeau AP, Laurell H. Estradiol accelerates endothelial healing through the retrograde commitment of uninjured endothelium. Am J Physiol Heart Circ Physiol 2008; 294:H2822-30. [PMID: 18441207 DOI: 10.1152/ajpheart.00129.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the accelerative effect of 17beta-estradiol (E2) on endothelial regrowth has been clearly demonstrated, the local cellular events accounting for this beneficial vascular action are still uncertain. In the present work, we compared the kinetics of endothelial healing of mouse carotid arteries after endovascular and perivascular injury. Both basal reendothelialization as well as the accelerative effect of E2 were similar in the two models. Three days after endothelial denudation, a regenerative area was observed in both models, characterized by similar changes in gene expression after injury, visualized by en face confocal microscopy (EFCM). A precise definition of the injury limits was only possible with the perivascular model, since it causes a complete and lasting decellularization of the media. Using this model, we demonstrated that the migration of uninjured endothelial cells precedes proliferation (bromodeoxyuridine incorporation) and that these events occur at earlier time points with E2 treatment. We have also identified an uninjured retrograde zone as an intimate component of the endothelial regeneration process. Thus, in the perivascular model, the regenerative area can be subdivided into a retrograde zone and a reendothelialized area. Importantly, both areas are significantly enlarged by E2. In conclusion, the combination of the electric perivascular injury model and EFCM is well adapted to the visualization of the endothelial monolayer and to investigate cellular events involved in reendothelialization. This process is accelerated by E2 as a consequence of the retrograde commitment of an uninjured endothelial zone to migrate and proliferate, contributing to an enlargement of the regenerative area.
Collapse
Affiliation(s)
- Cédric Filipe
- Institut National de la Santé et de la Recherche Médicale (INSERM) U858, I2MR, Equipe 9, Université de Toulouse III Paul Sabatier and Centre Hospitalier Universitaire de Toulouse-Rangueil, Toulouse, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lin CC, Yang WC, Lin SJ, Chen TW, Lee WS, Chang CF, Lee PC, Lee SD, Su TS, Fann CSJ, Chung MY. Length polymorphism in heme oxygenase-1 is associated with arteriovenous fistula patency in hemodialysis patients. Kidney Int 2006; 69:165-72. [PMID: 16374439 DOI: 10.1038/sj.ki.5000019] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, producing carbon monoxide (CO), which carries potent antiproliferative and anti-inflammatory effects in the vascular walls. Transcription of the HO-1 gene is regulated by the length polymorphism of dinucleotide guanosine thymine repeat (GT)(n) in the promoter region, which was measured in this study to determine its association with arteriovenous fistula (AVF) failure in Chinese hemodialysis (HD) patients in Taiwan. L allele means (GT)(n)>or=30 and S allele means (GT)n<30. Therefore, there are two L alleles for L/L genotype, one L and one S allele for L/S genotype, and two S alleles for S/S genotype. Among the 603 HD patients who were enrolled in this study, 178 patients had history of AVF failure, while 425 patients did not. Significant associations were found between AVF failure and the following factors (hazard ratio): longer HD duration (1.004 month), lower pump flow (0.993 ml/min), higher dynamic venous pressure (1.010 mmHg), location of AVF on the right side (1.587 vs left side) and upper arm (2.242 vs forearm), and L/L and L/S genotypes of HO-1 (2.040 vs S/S genotype). The proportion of AVF failure increased from 20.3% in S/S genotype and 31.0% in L/S genotype to 35.4% in L/L genotype (P=0.011). Relative incidences were 1/87.6 (1 episode per 87.6 patient-months), 1/129, and 1/224.9 for HD patients with L/L, L/S, and S/S genotypes, respectively (P<0.002). The unassisted patency of AVF at 5 years decreased significantly from 83.8% (124/148) to 75.1% (223/297) and 69% (109/158) in S/S, L/S, and L/L genotypes, respectively (P<0.0001). In comparison with HD patients with S/S genotype, those with L/L genotype had a higher prevalence of coronary artery disease (29.1 vs 14.2%; P=0.005). A longer length polymorphism with (GT)(n) >or=30 in the HO-1 gene was associated with a higher frequency of access failure and a poorer patency of AVF in HD patients. The longer GT repeat in the HO-1 promoter might inhibit gene transcription, and consequently offset the CO-mediated protective effect against vascular injury.
Collapse
Affiliation(s)
- C-C Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Dibra A, Kastrati A, Mehilli J, Pache J, von Oepen R, Dirschinger J, Schömig A. Influence of stent surface topography on the outcomes of patients undergoing coronary stenting: A randomized double-blind controlled trial. Catheter Cardiovasc Interv 2005; 65:374-80. [PMID: 15926182 DOI: 10.1002/ccd.20400] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The objective of this study was to examine the relationship between stent surface topography and outcome in patients undergoing implantation of stents with rough and smooth surfaces. Surface topography is considered an important determinant of the bare stent performance. Specifically designed rough surface may increase the drug-storing capacity of stents but its direct impact on the risk of thrombosis and restenosis is not known. A total of 200 patients with significant stenosis in native coronary vessels were randomly assigned in a double-blind way to receive either a rough or a smooth-surface stent. The primary endpoint of the study was late lumen loss. Secondary endpoints included angiographic restenosis and clinical outcomes. The study was designed to test the equivalence of rough-surface stents to smooth-surface stents with respect to late lumen loss based on a noninferiority margin of 0.20 mm. Follow-up angiography was performed in 77% of the patients. Late lumen loss was 1.0 +/- 0.7 mm in the rough-surface stent group and 1.2 +/- 0.7 mm in the smooth stent surface group with a mean difference of -0.20 mm (95% CI = -0.43 to 0.02) between the two stents (P < 0.001 from test for equivalence and P = 0.08 from test for superiority). Angiographic restenosis rates were 25% with rough-surface stents and 35% with smooth-surface stents (P = 0.19). These results show that a rough stent surface does not increase late lumen loss after stent implantation as compared with a conventional smooth stent surface.
Collapse
|
38
|
Abstract
Strategies that enhance the number of endothelial cells (ECs) in the vessel wall following injury may limit complications such as thrombosis, vasospasm, and neointimal formation through reconstitution of a luminal barrier and cellular secretion of paracrine factors. Proof of principle has been demonstrated by studies in which mature ECs, culture expanded from harvested vascular tissue, were seeded in the arterial wall following balloon injury. The recent identification of circulating cells capable of developing an endothelial phenotype, including progenitor cells, has raised the possibility of using blood-derived cells as therapeutic agents. This article reviews data suggesting that such cells confer vascular protective effects after injury, raising the potential for novel, autologous approaches to the treatment of vascular disease.
Collapse
Affiliation(s)
- Rajiv Gulati
- Department of Cardiovascular Medicine, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
39
|
Van Belle E, Rosey G, Bauters C, Lablanche JM. [Restenosis and vascular response after angioplasty: physiopathological data]. PATHOLOGIE-BIOLOGIE 2004; 52:176-9. [PMID: 15145129 DOI: 10.1016/j.patbio.2004.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Accepted: 01/28/2004] [Indexed: 04/29/2023]
Abstract
Our purpose is to review the different mechanisms involved in the vascular response following percutaneous coronary revascularization, and their role in thrombotic complications and in restenosis. Specificities related to the various techniques of percutaneous coronary revascularization (balloon angioplasty, coronary stents, brachytherapy, drug eluting stents) are discussed.
Collapse
Affiliation(s)
- E Van Belle
- Service de cardiologie B et hémodynamique, Hôpital cardiologique, boulevard du Professeur-J.-Leclerc, 59037 Lille, France.
| | | | | | | |
Collapse
|
40
|
Durante W. Heme oxygenase-1 in growth control and its clinical application to vascular disease. J Cell Physiol 2003; 195:373-82. [PMID: 12704646 DOI: 10.1002/jcp.10274] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Biliverdin is subsequently metabolized to bilirubin by the enzyme biliverdin reductase. Although interest in HO-1 originally centered on its heme-degrading function, recent findings indicate that HO-1 exerts other biologically important actions. Emerging evidence suggests that HO-1 plays a critical role in growth regulation. Deletion of the HO-1 gene or inhibition of HO-1 activity results in growth retardation and impaired fetal development, whereas HO-1 overexpression increases body size. Although the mechanisms responsible for the growth promoting properties of HO-1 are not well established, HO-1 can indirectly influence growth by regulating the synthesis of growth factors and by modulating the delivery of oxygen or nutrients to specific target tissues. In addition, HO-1 exerts important effects on critical determinants of tissue size, including cell proliferation, apoptosis, and hypertrophy. However, the actions of HO-1 are highly variable and may reflect a role for HO-1 in maintaining tissue homeostasis. Considerable evidence supports a crucial role for HO-1 in blocking the growth of vascular smooth muscle cells (SMCs). This antiproliferative effect of HO-1 is mediated primarily via the release of CO, which inhibits vascular SMC growth via multiple pathways. Pharmacologic or genetic approaches targeting HO-1 or CO to the blood vessel wall may represent a promising, novel therapeutic approach in treating vascular proliferative disorders.
Collapse
Affiliation(s)
- William Durante
- Houston VA Medical Center and Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
41
|
Agata J, Zhang JJ, Chao J, Chao L. Adrenomedullin gene delivery inhibits neointima formation in rat artery after balloon angioplasty. REGULATORY PEPTIDES 2003; 112:115-20. [PMID: 12667632 DOI: 10.1016/s0167-0115(03)00029-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Adrenomedullin (AM) is a potent vasodilator expressed in tissues relevant to cardiovascular function. AM peptide has been shown to inhibit the proliferation and migration of vascular smooth muscle cells in vitro. However, the effect of AM on blood vessels after vascular injury in vivo has not been elucidated. In order to explore the potential roles of AM in vascular biology, we evaluated the effect of AM by local gene delivery on neointima formation in balloon-injured rat artery. Adenovirus carrying the human AM cDNA under the control of cytomegalovirus promoter/enhancer (Ad.CMV-hAM) was generated by homologous recombination. After delivery of Ad.CMV-hAM into rat left carotid artery, we identified the expression of human AM mRNA in the left carotid artery, but not in the right carotid artery, heart or kidney by reverse transcription-polymerase chain reaction (RT-PCR) followed by Southern blot analysis. Following local AM gene delivery, we observed a 51% reduction in intima/media ratio at the injured site as compared with that of control rats injected with the luciferase gene (n=7, P<0.01). AM gene transfer resulted in regeneration of endothelium as compared to the control. AM gene delivery significantly increased cGMP levels in balloon-injured arteries. These results indicate that AM contributes to reduction of neointima formation by promotion of re-endothelialization and inhibition of vascular smooth muscle cell proliferation via cGMP-dependent signaling pathway.
Collapse
Affiliation(s)
- Jun Agata
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Avenue, PO Box 250509, Charleston, SC 29425-2211, USA
| | | | | | | |
Collapse
|
42
|
Indolfi C, Torella D, Coppola C, Curcio A, Rodriguez F, Bilancio A, Leccia A, Arcucci O, Falco M, Leosco D, Chiariello M. Physical training increases eNOS vascular expression and activity and reduces restenosis after balloon angioplasty or arterial stenting in rats. Circ Res 2002; 91:1190-7. [PMID: 12480821 DOI: 10.1161/01.res.0000046233.94299.d6] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The effects of dynamic exercise on restenosis after vascular injury are still unknown. The consequences of balloon dilation-induced injury on neointimal hyperplasia, vascular negative remodeling, and reendothelialization were assessed in sedentary and trained rats. Ex vivo eNOS vascular expression and activity were investigated in carotid arteries isolated from sedentary and exercised rats. The in vivo effects of eNOS inhibition by L-NMMA on vessel wall after balloon dilation were evaluated in sedentary and exercised rats. We also investigated the effects of exercise on neointimal formation in a rat stent model of vascular injury. Compared with sedentary group, the arteries isolated from trained rats showed higher levels of eNOS protein expression and activity 7 days after balloon dilation. A significant reduction of both neointimal hyperplasia and negative remodeling was observed 14 days after balloon injury in trained compared with sedentary rats. Moreover, we demonstrated that exercise training produced accelerated reendothelialization of the balloon injured arterial segments compared with sedentary. L-NMMA administration eliminated the benefits of physical training on vessel wall after balloon dilation. Finally, a decrease of neointimal hyperplasia as well as of platelet aggregation was observed after stent deployment in trained rats compared with sedentary. In conclusion, physical exercise could favorably affect restenosis after balloon angioplasty and stenting. Increase in eNOS expression and activity might contribute to the potential beneficial effects of exercise on the vessel wall after vascular injury.
Collapse
MESH Headings
- Adenosine Diphosphate/pharmacology
- Angioplasty, Balloon/adverse effects
- Animals
- Carotid Stenosis/etiology
- Carotid Stenosis/pathology
- Carotid Stenosis/prevention & control
- Cell Division/physiology
- Disease Models, Animal
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/injuries
- Endothelium, Vascular/pathology
- Enzyme Activation/drug effects
- Enzyme Inhibitors/pharmacology
- Graft Occlusion, Vascular/etiology
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/prevention & control
- Hyperplasia/pathology
- Hyperplasia/prevention & control
- Immunoblotting
- Immunohistochemistry
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type III
- Physical Conditioning, Animal
- Physical Exertion
- Platelet Aggregation/drug effects
- Rats
- Rats, Wistar
- Stents/adverse effects
- Swimming/physiology
- Tunica Intima/drug effects
- Tunica Intima/enzymology
- Tunica Intima/injuries
- Tunica Intima/pathology
- omega-N-Methylarginine/pharmacology
Collapse
Affiliation(s)
- Ciro Indolfi
- Division of Cardiology, Magna Graecia University, Catanzaro, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Underwood PA, Bean PA, Gamble JR. Rate of endothelial expansion is controlled by cell:cell adhesion. Int J Biochem Cell Biol 2002; 34:55-69. [PMID: 11733185 DOI: 10.1016/s1357-2725(01)00100-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Procedures used to alleviate blood vessel occlusion result in varying degrees of damage to the vascular wall and endothelial denudation. The presence of intact, functioning endothelium is thought to be important in controlling smooth muscle cell growth, and limiting the intimal thickening which results from damage to the vessel wall. Recovery of the endothelium is commonly slow and incomplete, due in part to endothelial lateral cell:cell adhesion, which limits cell migration and proliferation. We have investigated the effect of fibroblast growth factor 2 and vascular/endothelial growth factor on the relationship between the temporal distribution of the junctional adhesion proteins, platelet/endothelial cell adhesion molecule, vascular/endothelial cadherin and plakoglobin, and cellular migration and proliferation in an in vitro model of endothelial expansion. We found that whereas cell:cell junctions were initially disturbed to similar extents by single applications of the growth factors, outward cell migration and proliferation rates were inversely correlated with the speed at which cell:cell junctions were re-established. This occurred very rapidly with vascular/endothelial growth factor treatment and more slowly with fibroblast growth factor-2, resulting in more extensive outward migration and proliferation in response to the latter. Platelet/endothelial cell adhesion molecule and vascular/endothelial cadherin appeared to be associated with cell:cell junctional control of migration and proliferation, while plakoglobin did not contribute. It was concluded that the rate of endothelial expansion in response to growth factors, is limited by the rate of re-association of junctional complexes following initial disruption.
Collapse
Affiliation(s)
- P Anne Underwood
- CSIRO Molecular Science, P.O. Box 184, North Ryde, NSW 1670, Australia.
| | | | | |
Collapse
|
44
|
de Crom R, Wulf P, van Nimwegen H, Kutryk MJ, Visser P, van der Kamp A, Hamming J. Irradiated versus nonirradiated endothelial cells: effect on proliferation of vascular smooth muscle cells. J Vasc Interv Radiol 2001; 12:855-61. [PMID: 11435542 DOI: 10.1016/s1051-0443(07)61511-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Endovascular radiation therapy is a promising strategy for the prevention of restenosis. Radiation prevents proliferation of vascular smooth muscle cells, thereby reducing the incidence of restenosis, but may also affect the remaining endothelial cells. For this reason, a comparison was made between irradiated and nonirradiated endothelial cells and their effects on the proliferation of vascular smooth muscle cells in a coculture system was evaluated. MATERIALS AND METHODS A coculture system was used, in which both endothelial cells and vascular smooth muscle cells were grown on opposite sides of a semipermeable membrane. After a period of growth arrest, the proliferation of vascular smooth muscle cells was measured during four subsequent days. RESULTS The presence of endothelial cells stimulated the proliferation of vascular smooth muscle cells during the first days of analysis but had an inhibitory effect during the subsequent days (P <.5). gamma-irradiation of endothelial cells resulted in a complete blockage of the proliferation of these cells. However, irradiated endothelial cells affected the proliferation of vascular smooth muscle cells in coculture in a fashion comparable to nonirradiated endothelial cells (P >.5). CONCLUSION The results suggest that, in endovascular radiation therapy, irradiation of endothelial cells does not change their effects on the proliferative behavior of vascular smooth muscle cells.
Collapse
Affiliation(s)
- R de Crom
- Medical Genetics Centre, Department of Cell Biology and Genetics, Erasmus University, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Very limited success has been demonstrated with systemic pharmacological treatment to reduce the incidence of restenosis following angioplasty in patients. The lack of success of many of the pharmacotherapeutic agents in reducing the restenosis rates post-angioplasty and following stent implementation is believed to arise from inadequate concentrations of the agents at the lesion site. This has led to the development of various local delivery devices that would ideally deliver and retain adequate amounts of drug to the vessel wall for sufficient periods of time to ensure a therapeutic effect without inducing further injury or compromising blood flow. Local dosing would avoid systemic toxicity, and the use of modified balloon catheters or coated stents might enable percutaneous approaches.
Collapse
Affiliation(s)
- D S Ettenson
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | |
Collapse
|
46
|
Fenton M, Barker S, Kurz DJ, Erusalimsky JD. Cellular senescence after single and repeated balloon catheter denudations of rabbit carotid arteries. Arterioscler Thromb Vasc Biol 2001; 21:220-6. [PMID: 11156856 DOI: 10.1161/01.atv.21.2.220] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The hypothesis that increased cellular proliferation in the vasculature may lead to replicative senescence has been tested in a model of neointima formation. We have used a biomarker of replicative senescence, senescence-associated beta-galactosidase (SA-beta-gal), to detect senescence in rabbit carotid arteries subjected to single and double balloon denudations. We found an accumulation of senescent cells in the neointima and media of all injured vessels, in contrast to the near absence of such cells in control vessels. The relative area occupied by SA-beta-gal-positive cells was higher in vessels subjected to double denudation than in those subjected to single denudation, both in the neointima (0.99% versus 0.06%, respectively; P:<0.001) and in the media (0.11% versus 0.01%, respectively; P:<0.02). The majority of SA-beta-gal-positive cells were vascular smooth muscle cells, and a minority were endothelial cells. SA-beta-gal-positive cells showed no evidence of apoptosis by use of terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling. Our results indicate that the proliferative response that follows intraluminal injury to the artery leads to the emergence of senescent endothelial and smooth muscle cells. The demonstration that vascular cell senescence can occur in vivo suggests that this process may be involved in cardiovascular pathologies that have a proliferative component.
Collapse
Affiliation(s)
- M Fenton
- Cell Biology Group, Centre for Cardiovascular Biology and Medicine, Department of Medicine, Royal Free and University College Medical School, University College London, London, UK
| | | | | | | |
Collapse
|
47
|
Martínez-González J, Badimon L. [Reendotelization, intimal thickening and vascular remodeling. A common denominator?]. Rev Esp Cardiol 2000; 53:1425-7. [PMID: 11083998 DOI: 10.1016/s0300-8932(00)75258-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Hou D, Rogers PI, Toleikis PM, Hunter W, March KL. Intrapericardial paclitaxel delivery inhibits neointimal proliferation and promotes arterial enlargement after porcine coronary overstretch. Circulation 2000; 102:1575-81. [PMID: 11004150 DOI: 10.1161/01.cir.102.13.1575] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Catheter-based intrapericardial (IPC) delivery of therapeutic agents has recently been demonstrated. Paclitaxel is known to inhibit vascular smooth muscle cell proliferation. This study examined the effect of IPC instillation of paclitaxel on neointimal proliferation after balloon overstretch of porcine coronary arteries. METHODS AND RESULTS Overstretch injury of coronary arteries was followed by IPC administration of micellar paclitaxel at low dose (LD, 10 mg; n=6) or high dose (HD, 50 mg; n=7) or of control micelles (50 mg, n=5). Animals were euthanized 28 days after balloon dilation. Arterial injury indices were no different among the groups. The neointimal area, maximal intimal thickness, and adventitial thickness were significantly reduced in both LD (0.47+/-0.04 mm(2), 0.43+/-0.03 mm, and 0.35+/-0.02 mm, respectively) and HD (0.51+/-0.06 mm(2), 0.42+/-0.03 mm, and 0. 38+/-0.03 mm, respectively) paclitaxel groups compared with the control group (0.79+/-0.07 mm(2), 0.56+/-0.02 mm, and 0.47+/-0.02 mm, respectively; P:<0.001). Meanwhile, the vessel circumference measured at the external elastic lamina of paclitaxel-treated vessels was significantly larger than the control circumference. Apoptotic cells were found in the neointima. The apoptotic cell percentage was not different between the control (1.72%) and LD (2. 31%) groups but was higher in the HD group (7.07%, P:<0.0001 versus control and LD groups). Immunostaining for matrix metalloproteinase-2 revealed concurrent reduction in the HD group compared with the control and LD groups. CONCLUSIONS IPC space delivery of a single dose of paclitaxel significantly reduces vessel narrowing in this balloon-overstretch model. This effect is mediated by reduction of neointimal mass as well as positive vascular remodeling.
Collapse
Affiliation(s)
- D Hou
- Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | |
Collapse
|
49
|
Agata J, Miao RQ, Yayama K, Chao L, Chao J. Bradykinin B(1) receptor mediates inhibition of neointima formation in rat artery after balloon angioplasty. Hypertension 2000; 36:364-70. [PMID: 10988266 DOI: 10.1161/01.hyp.36.3.364] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We evaluated the effects of the kallikrein-kinin system on the proliferation and migration of primary cultured vascular smooth muscle cells (VSMCs) in vitro and neointima formation in balloon-injured rat carotid arteries in vivo. In cultured rat VSMCs, tissue kallikrein inhibited cell proliferation, and this inhibitory effect was blocked by Sar-Tyr-Aca(epsilon)-Lys [D-betaNal(7), Ile(8)]-des-Arg(9)-bradykinin, a bradykinin B(1) receptor antagonist, and by icatibant, a bradykinin B(2) receptor antagonist. Platelet-derived growth factor significantly increased the expression of the B(1) receptor but not the B(2) receptor in VSMCs. Platelet-derived growth factor-induced cell migration was significantly attenuated by des-Arg(9)-bradykinin and to a lesser degree by bradykinin. Endogenous B(1) receptor mRNA increased in rat carotid arteries after balloon angioplasty. After local delivery of adenovirus carrying the human tissue kallikrein gene into the rat carotid artery, we observed a 54% reduction in the intima/media ratio at the injured site compared with the control ratio (n=7, P:<0.01). Administration of the B(1) receptor antagonist via minipumps blocked the protective effect of kallikrein and partially reversed the intima/media ratio toward the control ratio. Kallikrein gene delivery results in the regeneration of endothelium compared with the control groups, and the B(1) receptor antagonist abolished this effect. Nitrite/nitrate, cGMP, and cAMP levels in balloon-injured arteries significantly increased after kallikrein gene delivery, whereas the B(1) receptor antagonist abolished these increases (n=4 or 5, P:<0.05). These results indicate that the B(1) receptor contributes to the reduction of neointima formation via the promotion of reendothelialization and inhibition of VSMC proliferation and migration through NO-cGMP and cAMP signaling pathways. This study provides significant implications in treating restenosis after revascularization.
Collapse
Affiliation(s)
- J Agata
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston 29425-2211, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
Advances in the field of molecular medicine are making gene therapy a viable treatment strategy for the next millennium. Indeed, over the past 10 years, a number of improvements have occurred that have resulted in an increased interest in gene therapy for the treatment of diseases in cardiovascular medicine. Because antithrombotic and anticoagulation therapy generally involves the systemic administration of agents that target a small region of the vasculature, localized and controlled delivery of specific genes could offer enormous potential to treat a number of life-threatening diseases. In addition, gene therapy may allow sustained antithrombotic or anticoagulant treatment when prolonged systemic administration is undesirable. Gene therapy for antithrombotic strategies can involve a number of different approaches. This could include inhibition of coagulation factors, over-expression of anticoagulant factors, or modulation of endothelial biology to make thrombus formation or propagation unfavorable. Preclinical data regarding these different strategies are reviewed and their potential limitations discussed.
Collapse
Affiliation(s)
- K M Channon
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | |
Collapse
|