1
|
Padmanaban AM, Ganesan K, Ramkumar KM. A Co-Culture System for Studying Cellular Interactions in Vascular Disease. Bioengineering (Basel) 2024; 11:1090. [PMID: 39593750 PMCID: PMC11591305 DOI: 10.3390/bioengineering11111090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are leading causes of morbidity and mortality globally, characterized by complications such as heart failure, atherosclerosis, and coronary artery disease. The vascular endothelium, forming the inner lining of blood vessels, plays a pivotal role in maintaining vascular homeostasis. The dysfunction of endothelial cells contributes significantly to the progression of CVDs, particularly through impaired cellular communication and paracrine signaling with other cell types, such as smooth muscle cells and macrophages. In recent years, co-culture systems have emerged as advanced in vitro models for investigating these interactions and mimicking the pathological environment of CVDs. This review provides an in-depth analysis of co-culture models that explore endothelial cell dysfunction and the role of cellular interactions in the development of vascular diseases. It summarizes recent advancements in multicellular co-culture models, their physiological and therapeutic relevance, and the insights they provide into the molecular mechanisms underlying CVDs. Additionally, we evaluate the advantages and limitations of these models, offering perspectives on how they can be utilized for the development of novel therapeutic strategies and drug testing in cardiovascular research.
Collapse
Affiliation(s)
- Abirami M. Padmanaban
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| |
Collapse
|
2
|
Balavigneswaran CK, Selvaraj S, Vasudha TK, Iniyan S, Muthuvijayan V. Tissue engineered skin substitutes: A comprehensive review of basic design, fabrication using 3D printing, recent advances and challenges. BIOMATERIALS ADVANCES 2023; 153:213570. [PMID: 37540939 DOI: 10.1016/j.bioadv.2023.213570] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/08/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
The multi-layered skin structure includes the epidermis, dermis and hypodermis, which forms a sophisticated tissue composed of extracellular matrix (ECM). The wound repair is a well-orchestrated process when the skin is injured. However, this natural wound repair will be ineffective for large surface area wounds. Autografts-based treatment is efficient but, additional pain and secondary healing of the patient limits its successful application. Therefore, there is a substantial need for fabricating tissue-engineered skin constructs. The development of a successful skin graft requires a fundamental understanding of the natural skin and its healing process, as well as design criteria for selecting a biopolymer and an appropriate fabrication technique. Further, the fabrication of an appropriate skin graft needs to meet physicochemical, mechanical, and biological properties equivalent to the natural skin. Advanced 3D bioprinting provides spatial control of the placement of functional components, such as biopolymers with living cells, which can satisfy the prerequisites for the preparation of an ideal skin graft. In this view, here we elaborate on the basic design requirements, constraints involved in the fabrication of skin graft and choice of ink, the probable solution by 3D bioprinting technique, as well as their latest advancements, challenges, and prospects.
Collapse
Affiliation(s)
- Chelladurai Karthikeyan Balavigneswaran
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India.
| | - Sowmya Selvaraj
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - T K Vasudha
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - Saravanakumar Iniyan
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - Vignesh Muthuvijayan
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India.
| |
Collapse
|
3
|
Mayer K, Ndrepepa G, Schroeter M, Emmer C, Bernlochner I, Schüpke S, Gewalt S, Hilz R, Coughlan JJ, Aytekin A, Heyken C, Morath T, Schunkert H, Laugwitz KL, Sibbing D, Kastrati A. High on-aspirin treatment platelet reactivity and restenosis after percutaneous coronary intervention: results of the Intracoronary Stenting and Antithrombotic Regimen-ASpirin and Platelet Inhibition (ISAR-ASPI) Registry. Clin Res Cardiol 2023; 112:1231-1239. [PMID: 36786829 PMCID: PMC10449652 DOI: 10.1007/s00392-023-02161-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/12/2023] [Indexed: 02/15/2023]
Abstract
OBJECTIVE The aim of this study was to assess the association between high on-aspirin treatment platelet reactivity (HAPR) and the subsequent risk of restenosis after percutaneous coronary intervention (PCI) with predominantly drug-eluting stents. BACKGROUND The association between HAPR and subsequent risk of restenosis after PCI is unclear. METHODS This study included 4839 patients undergoing PCI (02/2007-12/2011) in the setting of the Intracoronary Stenting and Antithrombotic Regimen-ASpirin and Platelet Inhibition (ISAR-ASPI) registry. Platelet function was assessed with impedance aggregometry using the multi-plate analyzer immediately before PCI and after intravenous administration of aspirin (500 mg). The primary outcome was clinical restenosis, defined as target lesion revascularization at 1 year. Secondary outcomes included binary angiographic restenosis and late lumen loss at 6- to 8-month angiography. RESULTS The upper quintile cut-off of platelet reactivity measurements (191 AU × min) was used to categorize patients into a group with HAPR (platelet reactivity > 191 AU × min; n = 952) and a group without HAPR (platelet reactivity ≤ 191 AU × min; n = 3887). The primary outcome occurred in 94 patients in the HAPR group and 405 patients without HAPR (cumulative incidence, 9.9% and 10.4%; HR = 0.96, 95% CI 0.77-1.19; P = 0.70). Follow-up angiography was performed in 73.2% of patients. There was no difference in binary restenosis (15.2% vs. 14.9%; P = 0.79) or late lumen loss (0.32 ± 0.57 vs. 0.32 ± 0.59 mm; P = 0.93) between patients with HAPR versus those without HAPR. CONCLUSIONS This study did not find an association between HAPR, measured at the time of PCI, and clinical restenosis at 1 year after PCI.
Collapse
Affiliation(s)
- Katharina Mayer
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Gjin Ndrepepa
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Mira Schroeter
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Christopher Emmer
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Isabell Bernlochner
- Medizinische Klinik and Poliklinik Innere Medizin I (Kardiologie, Angiologie, Pneumologie), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefanie Schüpke
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Senta Gewalt
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Raphaela Hilz
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - John Joseph Coughlan
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Alp Aytekin
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Clarissa Heyken
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Tanja Morath
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- Medizinische Klinik and Poliklinik Innere Medizin I (Kardiologie, Angiologie, Pneumologie), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Dirk Sibbing
- Klinik der Universität München, Cardiology, Ludwig-Maximilians-Universität, Munich, Germany
- Privatklinik Lauterbacher Mühle am Ostersee, Iffeldorf und Ludwig-Maximilians-Univerität, Munich, Germany
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Cardiology and Technische Universität München, Lazarettstr. 36, 80636, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
4
|
Kottmann P, Eildermann K, Murthi SR, Cleuziou J, Lemmer J, Vitanova K, von Stumm M, Lehmann L, Hörer J, Ewert P, Sigler M, Lange R, Lahm H, Dreßen M, Lichtner P, Wolf CM. EGFR and MMP-9 are associated with neointimal hyperplasia in systemic-to-pulmonary shunts in children with complex cyanotic heart disease. Mamm Genome 2023; 34:285-297. [PMID: 36867212 PMCID: PMC10290590 DOI: 10.1007/s00335-023-09982-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023]
Abstract
Systemic-to-pulmonary shunt malfunction contributes to morbidity in children with complex congenital heart disease after palliative procedure. Neointimal hyperplasia might play a role in the pathogenesis increasing risk for shunt obstruction. The aim was to evaluate the role of epidermal growth factor receptor (EGFR) and matrix-metalloproteinase 9 (MMP-9) in the formation of neointimal within shunts. Immunohistochemistry was performed with anti-EGFR and anti-MMP-9 on shunts removed at follow-up palliative or corrective procedure. Whole-genome single-nucleotide polymorphisms genotyping was performed on DNA extracted from patients´ blood samples and allele frequencies were compared between the group of patients with shunts displaying severe stenosis (≥ 40% of lumen) and the remaining group. Immunohistochemistry detected EGFR and MMP-9 in 24 of 31 shunts, located mainly in the luminal area. Cross-sectional area of EGFR and MMP-9 measured in median 0.19 mm2 (IQR 0.1-0.3 mm2) and 0.04 mm2 (IQR 0.03-0.09 mm2), respectively, and correlated positively with the area of neointimal measured on histology (r = 0.729, p < 0.001 and r = 0.0479, p = 0.018, respectively). There was a trend of inverse correlation between the dose of acetylsalicylic acid and the degree of EGFR, but not MMP-9, expression within neointima. Certain alleles in epidermal growth factor (EGF) and tissue inhibitor of metalloproteinases 1 (TIMP-1) were associated with increased stenosis and neointimal hyperplasia within shunts. EGFR and MMP-9 contribute to neointimal proliferation in SP shunts of children with complex cyanotic heart disease. SP shunts from patients carrying certain risk alleles in the genes encoding for EGF and TIMP-1 displayed increased neointima.
Collapse
Affiliation(s)
- Philip Kottmann
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Lazarettstrasse 36, 80636, Munich, Germany
| | - Katja Eildermann
- Department of Pediatrics and Adolescent Medicine-Paediatric Cardiology, Intensive Care Medicine and Pneumology, University Medical Center, Goettingen, Germany
| | - Sarala Raj Murthi
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Lazarettstrasse 36, 80636, Munich, Germany
| | - Julie Cleuziou
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilian University Munich, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Julia Lemmer
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Lazarettstrasse 36, 80636, Munich, Germany
| | - Keti Vitanova
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Maria von Stumm
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilian University Munich, Munich, Germany
| | - Luisa Lehmann
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Lazarettstrasse 36, 80636, Munich, Germany
| | - Jürgen Hörer
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilian University Munich, Munich, Germany
| | - Peter Ewert
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Lazarettstrasse 36, 80636, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Matthias Sigler
- Department of Pediatrics and Adolescent Medicine-Paediatric Cardiology, Intensive Care Medicine and Pneumology, University Medical Center, Goettingen, Germany
| | - Rüdiger Lange
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Harald Lahm
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Martina Dreßen
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Centrum Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Cordula M Wolf
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine & Health, Lazarettstrasse 36, 80636, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
5
|
Meki M, El-Baz A, Sethu P, Giridharan G. Effects of Pulsatility on Arterial Endothelial and Smooth Muscle Cells. Cells Tissues Organs 2022; 212:272-284. [PMID: 35344966 PMCID: PMC10782761 DOI: 10.1159/000524317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/16/2022] [Indexed: 01/04/2023] Open
Abstract
Continuous flow ventricular assist device (CFVAD) support in advanced heart failure patients causes diminished pulsatility, which has been associated with adverse events including gastrointestinal bleeding, end organ failure, and arteriovenous malformation. Recently, pulsatility augmentation by pump speed modulation has been proposed as a means to minimize adverse events. Pulsatility primarily affects endothelial and smooth muscle cells in the vasculature. To study the effects of pulsatility and pulse modulation using CFVADs, we have developed a microfluidic co-culture model with human aortic endothelial (ECs) and smooth muscle cells (SMCs) that can replicate physiologic pressures, flows, shear stresses, and cyclical stretch. The effects of pulsatility and pulse frequency on ECs and SMCs were evaluated during (1) normal pulsatile flow (120/80 mmHg, 60 bpm), (2) diminished pulsatility (98/92 mmHg, 60 bpm), and (3) low cyclical frequency (115/80 mmHg, 30 bpm). Shear stresses were estimated using computational fluid dynamics (CFD) simulations. While average shear stresses (4.2 dynes/cm2) and flows (10.1 mL/min) were similar, the peak shear stresses for normal pulsatile flow (16.9 dynes/cm2) and low cyclic frequency (19.5 dynes/cm2) were higher compared to diminished pulsatility (6.45 dynes/cm2). ECs and SMCs demonstrated significantly lower cell size with diminished pulsatility compared to normal pulsatile flow. Low cyclical frequency resulted in normalization of EC cell size but not SMCs. SMCs size was higher with low frequency condition compared to diminished pulsatility but did not normalize to normal pulsatility condition. These results may suggest that pressure amplitude augmentation may have a greater effect in normalizing ECs, while both pressure amplitude and frequency may be required to normalize SMCs morphology. The co-culture model may be an ideal platform to study flow modulation strategies.
Collapse
Affiliation(s)
- Moustafa Meki
- Bioengineering, University of Louisville, Louisville, KY, USA
| | - Ayman El-Baz
- Bioengineering, University of Louisville, Louisville, KY, USA
| | - Palanaippan Sethu
- Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
6
|
Regulation of bFGF-induced effects on rat aortic smooth muscle cells by β3-adrenergic receptors. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100094. [PMID: 35300074 PMCID: PMC8920869 DOI: 10.1016/j.crphar.2022.100094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 01/08/2023] Open
Abstract
Background Basic fibroblast growth factor (bFGF)-mediated vascular smooth muscle cell (VSMC) proliferation and migration play an important role in vascular injury-induced neointima formation and subsequent vascular restenosis, a major event that hinders the long-term success of angioplasty. The function of β3-adrenergic receptors (β3-ARs) in vascular injury-induced neointima formation has not yet been defined. Objectives Our current study explored the possible role of β3-ARs in vascular injury-induced neointima formation by testing its effects on bFGF-induced VSMC migration and proliferation. Methods β3-AR expression in rat carotid arteries was examined at 14 days following a balloon catheter-induced injury. The effects of β3-AR activation on bFGF-induced rat aortic smooth muscle cell proliferation, migration, and signaling transduction (including extracellular-signal-regulated kinase/mitogen activated protein kinase, ERK/MAPK and Protein kinase B, AKT) were tested. Results We found that vascular injury induced upregulation of β3-ARs in neointima. Pretreatment of VSMCs with a selective β3-AR agonist, CL316,243 significantly potentiated bFGF-induced cell migration and proliferation, and ERK and AKT phosphorylation. Our results also revealed that suppressing phosphorylation of ERK and AKT blocked bFGF-induced cell migration and that inhibiting AKT phosphorylation reduced bFGF-mediated cell proliferation. Conclusion Our results suggest that activation of β3-ARs potentiates bFGF-mediated effects on VSMCs by enhancing bFGF-mediated ERK and AKT phosphorylation and that β3-ARs may play a role in vascular injury-induced neointima formation. β3-adrenergic receptor (β3-AR) expression was upregulated in the newly formed intima following rat carotid artery injury. Activation of β3-ARs potentiated bFGF-induced VSMC migration and proliferation and phosphorylation of ERK and/or AKT. Inhibition of ERK or AKT pathways decreased bFGF-induced cell migration. Inhibition of AKT pathway decreased bFGF-induced cell proliferation.
Collapse
|
7
|
Yasaka M, Yamaguchi T, Ogata J. Moyamoya Disease. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
8
|
Kearney KE, Wallner K, Kim M, Hira RS, Kim EY, Nakamura K, Parvathaneni U, Steinberg ZL, McCabe JM, Lombardi WL, Phillips ML, Don C. Intravascular coronary brachytherapy combined with a drug-coated balloon. Brachytherapy 2021; 20:1276-1281. [PMID: 34226148 DOI: 10.1016/j.brachy.2021.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/30/2021] [Accepted: 04/19/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Coronary artery disease leads to stenosis of the major cardiac vessels, resulting in ischemia and infarction. Percutaneous intervention (PCI) with balloon angioplasty can re-open stenosed vessels. Drug eluting stents (DES) and intravascular brachytherapy (IVBT) and drug-coated balloons (DCBs) are proven to decrease the likelihood of another restenosis after PCI, but neither is completely effective. Due to the limited long-term effectiveness of IVBT or DCB used separately for salvage PCI, we combined the two in some poor prognosis patients. METHODS Combined IVBT+DCB was intended for a total of 36 patients from 2015-2020. PCI with some combination of ballooning, laser and directional/rotational atherectomy was used to maximally open the stenotic region prior to IVBT+DCB. Beta-radiation brachytherapy for all patients was done with a Novoste Beta-Cath. Lutonix 4.0 x 40 mm paclitaxel-coated balloons (Bard, Murray Hill, NJ) were employed. RESULTS Overall survival at two years was 88%. Nine patients had follow-up angiograms, all for cardiac symptoms. Time from IVBT+DCB to follow-up angiography ranged from 4 to 33 months. The average months PCI-free interval before brachy therapy was 11.1 mos (95% CI 1.03-23.25) versus 23.3 mos after VBT (23.3 95% CI 12.3-32.3). The mean difference was 11.2 mos (95% CI 1.06-21.4, p < 0.031). None of the follow-up angiographic procedures displayed evidence of what could be interpreted as radiation damage. CONCLUSIONS In this uncontrolled series, IVBT plus DCB appeared to lengthen the ISR-free interval relative to what had been achieved prior to the combined intervention. We view these results as mildly encouraging, worthy of further study.
Collapse
Affiliation(s)
| | - Kent Wallner
- Radiation Oncology, University of Washington, Seattle, WA.
| | - Minsun Kim
- Radiation Oncology, University of Washington, Seattle, WA
| | - Ravi S Hira
- Departments of Cardiology, University of Washington, Seattle, WA
| | - Edward Y Kim
- Radiation Oncology, University of Washington, Seattle, WA
| | - Kenta Nakamura
- Departments of Cardiology, University of Washington, Seattle, WA
| | | | | | - James M McCabe
- Departments of Cardiology, University of Washington, Seattle, WA
| | | | | | - Creighton Don
- Radiation Oncology, University of Washington, Seattle, WA
| |
Collapse
|
9
|
Seiffert N, Tang P, Keshi E, Reutzel-Selke A, Moosburner S, Everwien H, Wulsten D, Napierala H, Pratschke J, Sauer IM, Hillebrandt KH, Struecker B. In vitro recellularization of decellularized bovine carotid arteries using human endothelial colony forming cells. J Biol Eng 2021; 15:15. [PMID: 33882982 PMCID: PMC8059238 DOI: 10.1186/s13036-021-00266-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 04/07/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Many patients suffering from peripheral arterial disease (PAD) are dependent on bypass surgery. However, in some patients no suitable replacements (i.e. autologous or prosthetic bypass grafts) are available. Advances have been made to develop autologous tissue engineered vascular grafts (TEVG) using endothelial colony forming cells (ECFC) obtained by peripheral blood draw in large animal trials. Clinical translation of this technique, however, still requires additional data for usability of isolated ECFC from high cardiovascular risk patients. Bovine carotid arteries (BCA) were decellularized using a combined SDS (sodium dodecyl sulfate) -free mechanical-osmotic-enzymatic-detergent approach to show the feasibility of xenogenous vessel decellularization. Decellularized BCA chips were seeded with human ECFC, isolated from a high cardiovascular risk patient group, suffering from diabetes, hypertension and/or chronic renal failure. ECFC were cultured alone or in coculture with rat or human mesenchymal stromal cells (rMSC/hMSC). Decellularized BCA chips were evaluated for biochemical, histological and mechanical properties. Successful isolation of ECFC and recellularization capabilities were analyzed by histology. RESULTS Decellularized BCA showed retained extracellular matrix (ECM) composition and mechanical properties upon cell removal. Isolation of ECFC from the intended target group was successfully performed (80% isolation efficiency). Isolated cells showed a typical ECFC-phenotype. Upon recellularization, co-seeding of patient-isolated ECFC with rMSC/hMSC and further incubation was successful for 14 (n = 9) and 23 (n = 5) days. Reendothelialization (rMSC) and partial reendothelialization (hMSC) was achieved. Seeded cells were CD31 and vWF positive, however, human cells were detectable for up to 14 days in xenogenic cell-culture only. Seeding of ECFC without rMSC was not successful. CONCLUSION Using our refined decellularization process we generated easily obtainable TEVG with retained ECM- and mechanical quality, serving as a platform to develop small-diameter (< 6 mm) TEVG. ECFC isolation from the cardiovascular risk target group is possible and sufficient. Survival of diabetic ECFC appears to be highly dependent on perivascular support by rMSC/hMSC under static conditions. ECFC survival was limited to 14 days post seeding.
Collapse
Affiliation(s)
- Nicolai Seiffert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Department for Trauma and Orthopedic Surgery, Vivantes-Hospital Spandau, Berlin, Germany
| | - Peter Tang
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Eriselda Keshi
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Anja Reutzel-Selke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Simon Moosburner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hannah Everwien
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dag Wulsten
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hendrik Napierala
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Johann Pratschke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Igor M Sauer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Karl H Hillebrandt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Surgery, Campus Charité Mitte
- Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program, Charitéplatz 1, 10117, Berlin, Germany
| | - Benjamin Struecker
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program, Charitéplatz 1, 10117, Berlin, Germany.,Department of General, Visceral and Transplant Surgery, University Hospital Münster, Münster, Germany
| |
Collapse
|
10
|
Che Man R, Sulaiman N, Ishak MF, Bt Hj Idrus R, Abdul Rahman MR, Yazid MD. The Effects of Pro-Inflammatory and Anti-Inflammatory Agents for the Suppression of Intimal Hyperplasia: An Evidence-Based Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217825. [PMID: 33114632 PMCID: PMC7672569 DOI: 10.3390/ijerph17217825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Anti-atherogenic therapy is crucial in halting the progression of inflammation-induced intimal hyperplasia. The aim of this concise review was to methodically assess the recent findings of the different approaches, mainly on the recruitment of chemokines and/or cytokine and its effects in combating the intimal hyperplasia caused by various risk factors. Pubmed and Scopus databases were searched, followed by article selection based on pre-set inclusion and exclusion criteria. The combination of keywords used were monocyte chemoattractant protein-1 OR MCP-1 OR TNF-alpha OR TNF-α AND hyperplasia OR intimal hyperplasia OR neointimal hyperplasia AND in vitro. These keywords combination was incorporated in the study and had successfully identified 77 articles, with 22 articles were acquired from Pubmed, whereas 55 articles were obtained from Scopus. However, after title screening, only twelve articles meet the requirements of defined inclusion criteria. We classified the data into 4 different approaches, i.e., utilisation of natural product, genetic manipulation and protein inhibition, targeted drugs in clinical setting, and chemokine and cytokines induction. Most of the articles are working on genetic manipulation targeted on specific pathway to inhibit the pro-inflammatory factors expression. We also found that the utilisation of chemokine- and cytokine-related treatments are emerging throughout the years. However, there is no study utilising the combination of approaches that might give a better outcome in combating intimal hyperplasia. Hopefully, this concise review will provide an insight regarding the usage of different novel approaches in halting the progression of intimal hyperplasia, which serves as a key factor for the development of atherosclerosis in cardiovascular disease.
Collapse
Affiliation(s)
- Rohaina Che Man
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Correspondence: ; Tel.: +603-9145-6995
| |
Collapse
|
11
|
Abstract
Revascularization surgeries such as coronary artery bypass grafting (CABG) are sometimes necessary to manage coronary heart disease (CHD). However, more than half of these surgeries fail within 10 years due to the development of intimal hyperplasia (IH) among others. The cytokine transforming growth factor-beta (TGFß) and its signaling components have been found to be upregulated in diseased or injured vessels, and to promote IH after grafting. Interventions that globally inhibit TGFß in CABG have yielded contrasting outcomes in in vitro and in vivo studies including clinical trials. With advances in molecular biology, it becomes clear that TGFß exhibits both protective and damaging roles, and only specific components such as some Smad-dependent TGFß signaling mediate vascular IH. The activin receptor-like kinase (ALK)-mediated Smad-dependent TGFß signaling pathways have been found to be activated in human vascular smooth muscle cells (VSMCs) following injury and in hyperplastic preimplantation vein grafts. It appears that focused targeting of TGFß pathway constitutes a promising therapeutic target to improve the outcome of CABG. This study dissects the role of TGFß pathway in CABG failure, with particular emphasis on the therapeutic potentials of specific targeting of Smad-dependent and ALK-mediated signaling.
Collapse
Affiliation(s)
- Marzuq A Ungogo
- Department of Veterinary Pharmacology and Toxicology, 58989Ahmadu Bello University, Zaria, Nigeria.,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
12
|
Perrotta I. The microscopic anatomy of endothelial cells in human atherosclerosis: Focus on ER and mitochondria. J Anat 2020; 237:1015-1025. [PMID: 32735733 DOI: 10.1111/joa.13281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022] Open
Abstract
Once regarded merely as a bland lipid storage disease consequence of aging, atherosclerosis is currently considered a slow and continuous inflammatory process (partially controllable by treatment) with complex etiology involving a multitude of genetic and environmental risk factors which ultimately result in the formation of the plaque. The vascular endothelium, a monolayer of endothelial cells (ECs), is an important regulatory "organ" critical for cardiovascular homeostasis in health which also contributes significantly to the pathomechanisms of several disease states, including atherosclerosis. Over the years, there has been evidence highlighting the central role of endoplasmic reticulum (ER) in the maintenance of endothelial function and perturbations in ER biology have been proposed to adversely affect a diverse range of endothelial functions. Of particular interest is the evidence that under certain pathophysiological circumstances, abnormal ER ultrastructure correlates with altered ER function and signaling and can contribute to cell injury and apoptosis. Therefore, the ultrastructural traits of ER membranes can have important implications not only for their functional bearings but also for the etiology and pathophysiology of diverse human disorders. With regard to atherosclerosis, the focus of ER research has been centered on the molecular signals originated from the ER to manage conditions of stress, leaving the fine structure of this organelle an almost unexplored (but promising) area of studies. There is, also, increasing evidence that mitochondrial dysfunction plays a critical role in promoting cell apoptosis, inflammation, and oxidative stress, thereby contributing to atheroma growth. It is within this context that the present study has been undertaken to investigate the microscopic architecture of ECs in human atherosclerosis and to determine whether the potential structural abnormalities of ER and mitochondria may play a central pathogenic role in atherogenesis or may merely reflect the condition of a tissue whose integrity has already been disturbed or destroyed. For this purpose, transmission electron microscopy (TEM) remains a powerful technique that can not only provide information about the ultrastructural state of cell organelles but also allow the correlation between different subcellular alterations indicative of a certain pathophysiological condition and cellular response. The present study expands the spectrum of ultrastructural defects known to exist in human atherosclerosis and suggests that ER alterations may be of great importance in the pathogenesis of the disease. The architectural changes of ER may be considered early pathological events that precede any overt histologic abnormalities in the vascular endothelium and its subcellular organelles, primarily the mitochondrial pool.
Collapse
Affiliation(s)
- Ida Perrotta
- Centre for Microscopy and Microanalysis, Transmission Electron Microscopy Laboratory, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
13
|
The quest for effective pharmacological suppression of neointimal hyperplasia. Curr Probl Surg 2020; 57:100807. [PMID: 32771085 DOI: 10.1016/j.cpsurg.2020.100807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
|
14
|
Chen P, He F, Liu T, Ma S, Shi B. Construction of calcitonin gene-related peptide-modified mesenchymal stem cells and analysis of their effects on the migration and proliferation of vascular smooth muscle cells. In Vitro Cell Dev Biol Anim 2020; 56:181-191. [PMID: 31912458 DOI: 10.1007/s11626-019-00429-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 12/09/2019] [Indexed: 10/25/2022]
Abstract
Lentiviral expression vectors for calcitonin gene-related peptide (CGRP) were used to transfect rat bone marrow mesenchymal stem cells (MSCs). After assessing the biological characteristics of proliferation and aging in MSCs transfected with CGRP, we observed the effects of the CGRP-modified rat MSCs on the migration and proliferation of rat vascular smooth muscle cells (VSMCs) in vitro. Rat MSCs were isolated, cultured in vitro, and identified by flow cytometry. A CGRP recombinant lentivirus was transfected into MSCs. The transfection efficiency was determined by fluorescence microscopy and flow cytometry, and CGRP in MSCs was detected by real-time quantitative PCR, ELISA, and immunofluorescence. The proliferation and senescence of CGRP-modified MSCs were evaluated by MTT assay and beta-galactosidase staining. VSMCs were isolated, cultured in vitro, and identified by immunofluorescence. CGRP-modified MSCs and VSMCs were cocultured in a Transwell system. The proliferation and migration of VSMCs were evaluated by scratch testing and the MTT method. Rat bone marrow MSCs showed a spindle-shaped morphology, adherent growth in vitro, positive CD29 and CD90 expression, and negative CD45 expression. CGRP was stably expressed in MSCs after 48 h of recombinant lentivirus transfection. CGRP mRNA and protein secretion in CGRP recombinant lentivirus-transfected MSCs were higher than that in control MSCs. Immunofluorescence showed that CGRP protein could be expressed in CGRP-modified MSCs. The proliferation ability and senescence rates did not differ between lentivirus-transfected MSCs and untransfected MSCs. Rat VSMCs expressed α-SMA protein and exhibited a spindle-shaped morphology and adherent growth in vitro. In Transwell coculture experiments, scratch testing of VSMCs showed that CGRP-modified MSCs could reduce VSMC proliferation and migration. The CGRP gene can be stably expressed in MSCs after CGRP recombinant lentivirus transfection. CGRP recombinant lentivirus transfection has little effect on the proliferation or senescence of MSCs, and CGRP-modified MSCs can inhibit the proliferation and migration of VSMCs. These results lay a foundation for research on the use of CGRP gene-engineered MSCs in restenosis therapy.
Collapse
Affiliation(s)
- Panke Chen
- Department of Cardiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Fang He
- Key Laboratory of Cell Engineering in Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Tao Liu
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Shuai Ma
- Department of Cardiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Bei Shi
- Department of Cardiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China.
| |
Collapse
|
15
|
Nies A, Proft L, Nehring ME, Gruber C, Sievers H, Hünigen H, Gemeinhardt O, Richardson KC, Hiebl B. Growth-related micromorphological characteristics of the porcine common carotid artery. Anat Histol Embryol 2019; 49:620-626. [PMID: 31797410 DOI: 10.1111/ahe.12522] [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: 07/24/2019] [Revised: 09/20/2019] [Accepted: 11/08/2019] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to gain knowledge about the micromorphology of the porcine common carotid artery (CCA) during the period of growth over the bodyweight range of 10-40 kg. CCA samples from German landrace pigs (DL) aged either 2 or 3 months (DL-2 and DL-3) were compared with samples from Göttingen minipigs (GM) aged either 18 or 40 months (GM-18 and GM-40) using transmitted light (phase-contrast mode) and transmission electron microscopy. The GM-18, GM-40 and the DL-3 groups had typical muscular artery histological characteristics. Contrasting to this, the 2-month-old DL pigs had a transitional artery type being characterized by a significantly higher proportion of elastic fibres and a significantly lower number of smooth muscle cells than did the 1 month older DL-3. During the period of maturation, the tunica media of the CCA in GM animals thickened by 1.3× and in DL animals by 2.5× resulting in an overall increased vessel wall thickness. The cumulated thickness of the tunica interna (endothelium, stratum subendotheliale and internal elastic lamina) and the tunica media (including the external elastic lamina) of DL-3 and GM-40 pigs were similar to each other and comparable to that of humans. With an increasing vessel wall thickness, the luminal diameter decreased in GM by 19% and in DL by 11%. Additionally, in the older age groups, GM-40 and DL-3, the internal elastic lamina principally was continuous, but there were also interrupted large segments of elastic lamina separated by gaps. In addition, the principal internal elastic lamina was duplicated in several places.
Collapse
Affiliation(s)
- Andrea Nies
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Lilly Proft
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Marie E Nehring
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Christian Gruber
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Henrieke Sievers
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Hana Hünigen
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Ole Gemeinhardt
- Corporate Member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Radiology, Berlin Institute of Health, Berlin, Germany
| | - Kenneth C Richardson
- School of Veterinary and Life Sciences, College of Veterinary Medicine, Murdoch University, Perth, WA, Australia
| | - Bernhard Hiebl
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| |
Collapse
|
16
|
Chen L, Pang S, Hao C, Xie A, Zhu K, He Y, Zhang X, Lu W, Ma G, Chen Z. Prevention of neointimal formation after angioplasty using tetramethylpyrazine-coated balloon catheters in a rabbit iliac artery model. Cardiovasc Diagn Ther 2019; 9:472-480. [PMID: 31737518 DOI: 10.21037/cdt.2019.09.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Restenosis remains a clinical problem; drug-coated balloons (DCBs) have demonstrated high efficiency in this situation. DCBs prevent neointimal hyperplasia by inhibiting cell proliferation and migration. Tetramethylpyrazine (TMP) is a traditional Chinese medicine originally isolated from the rhizome of Ligusticum Walliichii, which can inhibit platelet aggregation and smooth muscle cell proliferation. We hypothesized that TMP-coated balloons (TCB) could reduce neointimal hyperplasia through the NF-κB signalling pathway. Methods Twenty-one New-Zealand White rabbits (2.5-3.0 kg, male) were fed high-fat diets; 36 bilateral iliac artery stenosis models were successfully established by balloon straining. Rabbits were randomly treated with TCB (n=20) or plain balloons (PBA, n=16) (3 died during model construction). Angiographies were recorded at baseline, the immediate period, and 4 weeks later. Animals were euthanized and arteries collected for histological analysis and immunohistochemical staining. Protein expression of proliferating cell nuclear antigen (PCNA) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 of the vessel samples were analyzed using Western blotting. Results No difference existed in the baseline lesion characteristics or procedural results. Angiographic follow-up was successfully performed on 18 rabbits (TCB: n=20, PBA: n=16), except for 3 deaths related to the operation. Treatment with TCB was superior to that with PBA, with lower late lumen loss (0.45±0.23 vs. 0.84±0.17 mm, P<0.01). Pathological analysis confirmed the efficiency of TCB through decreasing the area stenosis rate compared with PBA (46.48%±8.22% vs. 75.24%±6.10%, P<0.01). As determined by Western blotting, significant reductions occurred in PCNA and NF-κB p65 protein intensity in the TCB group versus the PBA group (all P<0.01). TCB efficiently mitigated restenosis in the rabbit iliac artery model. Conclusions This study elucidated that TCB could restrain intimal hyperplasia of vessels by inhibiting the activation of the NF-κB pathway to reduce inflammatory response and decrease the rate of cell proliferation through suppressing PCNA expression.
Collapse
Affiliation(s)
- Lijuan Chen
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Si Pang
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Aiming Xie
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Kongbo Zhu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Yanru He
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Xiaoguo Zhang
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Wenbing Lu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Zhong Chen
- Department of Cardiology, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital East, Shanghai 201306, China.,Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
17
|
Homozygous receptors for insulin and not IGF-1 accelerate intimal hyperplasia in insulin resistance and diabetes. Nat Commun 2019; 10:4427. [PMID: 31562314 PMCID: PMC6765023 DOI: 10.1038/s41467-019-12368-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/03/2019] [Indexed: 11/08/2022] Open
Abstract
Insulin and IGF-1 actions in vascular smooth muscle cells (VSMC) are associated with accelerated arterial intima hyperplasia and restenosis after angioplasty, especially in diabetes. To distinguish their relative roles, we delete insulin receptor (SMIRKO) or IGF-1 receptor (SMIGF1RKO) in VSMC and in mice. Here we report that intima hyperplasia is attenuated in SMIRKO mice, but not in SMIGF1RKO mice. In VSMC, deleting IGF1R increases homodimers of IR, enhances insulin binding, stimulates p-Akt and proliferation, but deleting IR decreases responses to insulin and IGF-1. Studies using chimeras of IR(extracellular domain)/IGF1R(intracellular-domain) or IGF1R(extracellular domain)/IR(intracellular-domain) demonstrate homodimer IRα enhances insulin binding and signaling which is inhibited by IGF1Rα. RNA-seq identifies hyaluronan synthase2 as a target of homo-IR, with its expression increases by IR activation in SMIGF1RKO mice and decreases in SMIRKO mice. Enhanced intima hyperplasia in diabetes is mainly due to insulin signaling via homo-IR, associated with increased Has2 expression.
Collapse
|
18
|
Zhang W, Liu D, Han X, Ren J, Zhou P, Ding P. MicroRNA-451 inhibits vascular smooth muscle cell migration and intimal hyperplasia after vascular injury via Ywhaz/p38 MAPK pathway. Exp Cell Res 2019; 379:214-224. [PMID: 30930138 DOI: 10.1016/j.yexcr.2019.03.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/09/2023]
Abstract
Increasing evidence has indicated that intimal hyperplasia is a common event in the pathophysiology of many vascular diseases including atherosclerosis (AS). Recently, deregulated microRNAs (miRNAs) have been reported to be associated with the pathophysiology of AS. However, the biological function and regulatory mechanisms of miRNAs in intimal hyperplasia in AS remain largely unclear. The aim of this study was to investigate the effects of miRNAs on intimal hyperplasia and reveal the underlying mechanisms of their effects. Firstly, the model of rat vascular injury was successfully constructed in vivo. Then, the miRNAs expression profiles were analyzed by miRNA microarray. It was observed that miR-451 was significantly downregulated in injury carotid arteries. Subsequently, we investigated miR-451 function and found that upregulation of miR-451 by agomir-451 improves intimal thickening in rats following vascular injury. It was also observed that miR-451 was downregulated in the VSMCs following platelet-derived growth factor type BB (PDGF-BB) stimulation. The upregulation of miR-451 attenuated PDGF-BB-induced VSMCs injury, as evidenced by inhibition of proliferation, invasion and migration. Besides, overexpression of miR-451 blocked the activation of p38 MAPK signaling pathway in PDGF-BB treated VSMCs, as demonstrated by the downregulation of phosphorylated (p-) p38. In addition, Ywhaz, a positive regulator of p38 MAPK signaling pathway, was found to be a direct target of miR-451 in the VSMCs and this was validated using a luciferase reporter assay. Overexpression of Ywhaz partially abolished the inhibitory effects of miR-451 overexpression on PDGF-BB induced VSMCs injury. Collectively, these findings indicated that miR-451 protected intimal hyperplasia and PDGF-BB-induced VSMCs injury by Ywhaz/p38 MAPK pathway, and miR-451 may be considered as a potential therapeutic target in the treatment of AS.
Collapse
Affiliation(s)
- Wenguang Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dongmei Liu
- Department of Radiation Oncology, Henan Province Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jianzhuang Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Pengli Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Pengxu Ding
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
19
|
Ammann KR, DeCook KJ, Li M, Slepian MJ. Migration versus proliferation as contributor to in vitro wound healing of vascular endothelial and smooth muscle cells. Exp Cell Res 2019; 376:58-66. [PMID: 30660619 PMCID: PMC6988716 DOI: 10.1016/j.yexcr.2019.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/31/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022]
Abstract
Wound closure, as a result of collective cell growth, is an essential biological response to injury. In the field of vascular biology, the response of vascular smooth muscle cells (SMCs) and endothelial cells (ECs) to injury and substrate surface is important in therapeutic clinical treatment interventions such as angioplasty and atherectomy. Specifically, the mechanism by which cells close wounds (i.e. proliferation versus migration) in response to injury stimuli is of interest to better modulate recurrent vascular stenosis, prevent thrombus formation, occlusion, and life-threatening cardiovascular events. Here, we examine growth extent and temporal sequence of events following wound or gap introduction to a confluent monolayer of vascular SMCs or ECs. Significant differences in the preferred mechanisms of these cells to close wounds or gaps were observed; after 48 h, 73% of SMC wound closure was observed to be due to proliferation, while 75% of EC wound closure resulted from migration. These mechanisms were further modulated via addition or removal of extracellular matrix substrate and injury, with ECs more responsive to substrate composition and less to injury, in comparison to SMCs. Our results indicate that ECs and SMCs heal wounds differently, and that the time and mode of injury and associated substrate surface all impact this response.
Collapse
Affiliation(s)
- Kaitlyn R Ammann
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA
| | - Katrina J DeCook
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA
| | - Maxwell Li
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA
| | - Marvin J Slepian
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA; Department of Medicine, Sarver Heart Center, University of Arizona, 1501 N Campbell Ave, PO Box 245035, Tucson, AZ 85724, USA.
| |
Collapse
|
20
|
Kato Y, Yokoyama U, Fujita T, Umemura M, Kubota T, Ishikawa Y. Epac1 deficiency inhibits basic fibroblast growth factor-mediated vascular smooth muscle cell migration. J Physiol Sci 2019; 69:175-184. [PMID: 30084082 PMCID: PMC11117070 DOI: 10.1007/s12576-018-0631-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/24/2018] [Indexed: 01/12/2023]
Abstract
Vascular smooth muscle cell (VSMC) migration and the subsequent intimal thickening play roles in vascular restenosis. We previously reported that an exchange protein activated by cAMP 1 (Epac1) promotes platelet-derived growth factor (PDGF)-induced VSMC migration and intimal thickening. Because basic fibroblast growth factor (bFGF) also plays a pivotal role in restenosis, we examined whether Epac1 was involved in bFGF-mediated VSMC migration. bFGF-induced lamellipodia formation and migration were significantly decreased in VSMCs obtained from Epac1-/- mice compared to those in Epac1+/+-VSMCs. The bFGF-induced phosphorylation of Akt and glycogen synthase kinase 3β (GSK3β), which play a role in bFGF-induced cell migration, was attenuated in Epac1-/--VSMCs. Intimal thickening induced by the insertion of a large wire was attenuated in Epac1-/- mice, and was accompanied by the decreased phosphorylation of GSK3β. These data suggest that Epac1 deficiency attenuates bFGF-induced VSMC migration, possibly via Akt/GSK3β pathways.
Collapse
Affiliation(s)
- Yuko Kato
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
- Department of Immunopathology, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Tetsuo Kubota
- Department of Immunopathology, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
21
|
Yi B, Shen Y, Tang H, Wang X, Li B, Zhang Y. Stiffness of Aligned Fibers Regulates the Phenotypic Expression of Vascular Smooth Muscle Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:6867-6880. [PMID: 30676736 DOI: 10.1021/acsami.9b00293] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Electrospun uniaxially aligned ultrafine fibers show great promise in constructing vascular grafts mimicking the anisotropic architecture of native blood vessels. However, understanding how the stiffness of aligned fibers would impose influences on the functionality of vascular cells has yet to be explored. The present study aimed to explore the stiffness effects of electrospun aligned fibrous substrates (AFSs) on phenotypic modulation in vascular smooth muscle cells (SMCs). A stable jet coaxial electrospinning (SJCES) method was employed to generate highly aligned ultrafine fibers of poly(l-lactide- co-caprolactone)/poly(l-lactic acid) (PLCL/PLLA) in shell-core configuration with a remarkably varying stiffness region from 0.09 to 13.18 N/mm. We found that increasing AFS stiffness had no significant influence on the cellular shape and orientation along the fiber direction with the cultured human umbilical artery SMCs (huaSMCs) but inhibited the cell adhesion rate, promoted cell proliferation and migration, and especially enhanced the F-actin fiber assembly in the huaSMCs. Notably, higher fiber stiffness resulted in significant downregulation of contractile markers like alpha-smooth muscle actin (α-SMA), smooth muscle myosin heavy chain, calponin, and desmin, whereas upregulated the gene expression of pathosis-associated osteopontin ( OPN) in the huaSMCs. These results allude to the phenotype of huaSMCs on stiffer AFSs being miserably modulated into a proliferative and pathological state. Consequently, it adversely affected the proliferation and migration behavior of human umbilical vein endothelial cells as well. Moreover, stiffer AFSs also revealed to incur significant upregulation of inflammatory gene expression, such as interleukin-6 ( IL-6), monocyte chemoattractant protein-1 ( MCP-1), and intercellular adhesion molecule-1 ( ICAM-1), in the huaSMCs. This study stresses that although electrospun aligned fibers are capable of modulating native-like oriented cell morphology and even desired phenotype realization or transition, they might not always direct cells into correct functionality. The integrated fiber stiffness underlying is thereby a critical parameter to consider in engineering structurally anisotropic tissue-engineered vascular grafts to ultimately achieve long-term patency.
Collapse
Affiliation(s)
| | | | | | | | - Bin Li
- Department of Orthopaedics , The First Affiliated Hospital of Soochow University , Suzhou 215006 , China
- Orthopaedic Institute, Medical College , Soochow University , Suzhou 215007 , China
- China Orthopaedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| | - Yanzhong Zhang
- China Orthopaedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| |
Collapse
|
22
|
Javadzadegan A, Moshfegh A, Afrouzi HH, Omidi M. Magnetohydrodynamic blood flow in patients with coronary artery disease. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2018; 163:111-122. [PMID: 30119846 DOI: 10.1016/j.cmpb.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/26/2018] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVES We aim to investigate the effect of a magnetic field with varying intensities on haemodynamic perturbations in a cohort of patients with coronary artery disease. METHODS Transient computational fluid dynamics (CFD) simulations were performed in three-dimensional (3D) models of coronary arteries reconstructed from 3D quantitative coronary angiography. The effect of magnetic field on wall shear stress (WSS) derived parameters including maximum wall shear stress (MWSS) and size of regions with low wall shear stress (ALWSS) as well as length of flow recirculation zones were determined. RESULTS The results showed a substantial reduction in MWSS, ALWSS and length of flow recirculation zones in the presence of magnetic field, in particular for coronaries with moderate to severe stenoses. When the whole cohort examined, time-averaged wall shear stress (TAWSS), ALWSS and the length of flow recirculation zones in the absence of magnetic field were approximately 1.71, 4.69 and 8.46 times greater than those in the presence of magnetic field, respectively. CONCLUSION Our findings imply that an externally applied magnetic field can improve haemodynamic perturbations in human coronary arteries.
Collapse
Affiliation(s)
- Ashkan Javadzadegan
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; ANZAC Research Institute, The University of Sydney, Sydney, NSW 2139, Australia; Concord Repatriation General Hospital, Sydney Local Health District, NSW 2139, Australia.
| | - Abouzar Moshfegh
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; ANZAC Research Institute, The University of Sydney, Sydney, NSW 2139, Australia; Concord Repatriation General Hospital, Sydney Local Health District, NSW 2139, Australia
| | | | - Mohammad Omidi
- Faculty of Mechanical Engineering, Babol Noshirvani University of Technology, Babol, Iran
| |
Collapse
|
23
|
Javadzadegan A, Moshfegh A, Behnia M. Effect of magnetic field on haemodynamic perturbations in atherosclerotic coronary arteries. J Med Eng Technol 2018; 42:148-156. [DOI: 10.1080/03091902.2018.1447034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ashkan Javadzadegan
- Macquarie University, Sydney, Australia
- ANZAC Research Institute, The University of Sydney, Sydney, Australia
| | - Abouzar Moshfegh
- Macquarie University, Sydney, Australia
- ANZAC Research Institute, The University of Sydney, Sydney, Australia
| | - Masud Behnia
- School of Management, Macquarie University, Sydney, Australia
| |
Collapse
|
24
|
Fujimoto W, Sawada T, Toba T, Takahashi Y, Miyata T, Oishi S, Osue T, Onishi T, Takaya T, Shimane A, Taniguchi Y, Kawai H, Yasaka Y. Comparison of the 9-month intra-stent conditions and 2-year clinical outcomes after Resolute zotarolimus-eluting stent implantation between 3-month and standard dual antiplatelet therapy. J Cardiol 2018; 72:66-73. [PMID: 29456058 DOI: 10.1016/j.jjcc.2018.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/27/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND The use of short-duration dual antiplatelet therapy (DAPT) remains controversial. To investigate efficacy and safety of short-duration DAPT, we performed a detailed comparison of intra-stent conditions by optical coherence tomography (OCT) after second-generation drug-eluting stent implantation with short-term and standard DAPT. METHODS AND RESULTS Eighty-two consecutive patients with stable angina pectoris who received Resolute zotarolimus-eluting stents (R-ZESs; Medtronic Cardiovascular, Santa Rosa, CA, USA) were enrolled. Patients were assigned to 3-month (3M group: 41 patients) and standard (standard group: 41 patients) DAPT. In the 3M group, clopidogrel was discontinued 3 months after stent implantation. In the standard group, DAPT was maintained until follow-up OCT. At 9 months, neointimal proliferation was significantly larger in the 3M group, but there were no significant between-group differences in the proportion of uncovered and malapposed strut. The prevalence of abnormal intra-stent tissue (AIT) at 9 months was equivalent between groups. A multiple regression analysis revealed malapposition at 9 months as the strongest independent predictor of AIT at 9 months, and the prevalence of AIT was not associated with DAPT duration. Over 2 years, cardiac events were equal between groups; however, major bleeding was higher tendency in the standard group than in the 3M group. CONCLUSION This OCT study indicated that reducing DAPT's duration may provide acceptable arterial healing in patients with implanted R-ZESs.
Collapse
Affiliation(s)
- Wataru Fujimoto
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Takahiro Sawada
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan.
| | - Takayoshi Toba
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Yu Takahashi
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Taishi Miyata
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Shogo Oishi
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Tsuyoshi Osue
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Tetsuari Onishi
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Tomofumi Takaya
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Akira Shimane
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Yasuyo Taniguchi
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Hiroya Kawai
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| | - Yoshinori Yasaka
- Hyogo Prefectural Himeji Cardiovascular Center, Division of Cardiovascular Medicine, Himeji, Hyogo, Japan
| |
Collapse
|
25
|
He Y, Mei L, Jin Y, Li XP, Jin C. Overexpression of Hepatocyte Growth Factor mRNA Induced by Gene Transfer Attenuates Neointimal Hyperplasia After Balloon Injury. Hum Gene Ther 2018; 29:816-827. [PMID: 29382231 DOI: 10.1089/hum.2017.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hepatic growth factor (HGF) has been widely used in studies on arterial remodeling after injury, and results turn out to be inconsistent. The changes of endogenous HGF expression after injury also remain controversial. This study clarified the role of exogenous human HGF (hHGF) gene transfer in neointimal hyperplasia and investigated the associated alterations of endogenous HGF and c-Met expressions under endothelial denudation with or without hHGF gene transfer using a balloon-injured rabbit aorta model. Sixty-one rabbits were randomly divided into normal controls, endothelial injury, endothelial injury with hHGF, or the control vector gene transfer groups. On weeks 1, 2, 4, and 8 after injury, neointimal hyperplasia and endothelialization were evaluated by the ratio of neointimal area to medial area (N/M ratio), CD31-positive staining, α-smooth muscle actin, and endothelial nitric oxide synthase expressions using histological analysis, immunohistochemistry staining, or real-time quantitative reverse transcriptase polymerase chain reaction. Endogenous rabbit HGF (rHGF) and c-Met expressions were detected with immunohistochemistry staining and quantitative reverse transcriptase polymerase chain reaction. It was found that expressions of endogeneous rHGF and c-Met in endothelial injury upregulated with peak levels on week 2 or week 4 after injury (p < 0.01). On week 1 after hHGF transfer, neointimal hyperplasia was significantly inhibited (p < 0.001), with decreased α-smooth muscle actin expression (p < 0.05) and improved endothelial cells regeneration and function (p < 0.01). More remarkable overexpression of endogenous rHGF and c-Met mRNAs were detected, and lowered positive staining of rHGF and c-Met was shown in the neointima (p < 0.05). These results demonstrated hHGF gene transfer induced further overexpression of endogenous rHGF and c-Met mRNAs but lowered immunoreactivities of rHGF and c-Met in the neointima, thus leading to significant attenuation of neointimal hyperplasia.
Collapse
Affiliation(s)
- Yu He
- 1 Department of Ultrasound, China-Japan Union Hospital of Jilin University , Changchun, China
| | - Li Mei
- 2 Department of Ultrasound, the First Hospital of Jilin University , Changchun, China
| | - Ying Jin
- 3 Department of Surgery, the First Hospital of Jilin University , Changchun, China
| | - Xiao-Ping Li
- 1 Department of Ultrasound, China-Japan Union Hospital of Jilin University , Changchun, China
| | - Chunxiang Jin
- 1 Department of Ultrasound, China-Japan Union Hospital of Jilin University , Changchun, China
| |
Collapse
|
26
|
Farahmand L, Darvishi B, Majidzadeh-A K. Suppression of chronic inflammation with engineered nanomaterials delivering nuclear factor κB transcription factor decoy oligodeoxynucleotides. Drug Deliv 2017; 24:1249-1261. [PMID: 28870118 PMCID: PMC8240980 DOI: 10.1080/10717544.2017.1370511] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As a prototypical pro-inflammatory transcription factor, constitutive activation of NF-κB signaling pathway has been reported in several chronic inflammatory disorders including inflammatory bowel disease, cystic fibrosis, rheumatoid arthritis and cancer. Application of decoy oligodeoxynucleotides (ODNs) against NF-κB, as an effective molecular therapy approach, has brought about several promising outcomes in treatment of chronic inflammatory disorders. However, systematic administration of these genetic constructs is mostly hampered due to their instability, rapid degradation by nucleases and poor cellular uptake. Both chemical modification and application of delivery systems have shown to effectively overcome some of these limitations. Among different administered delivery systems, nanomaterials have gained much attention for delivering NF-κB decoy ODNs owing to their high loading capacity, targeted delivery and ease of synthesis. In this review, we highlight some of the most recently developed nanomaterial-based delivery systems for overcoming limitations associated with clinical application of these genetic constructs.
Collapse
Affiliation(s)
- Leila Farahmand
- a Recombinant Proteins Department , Motamed Breast Cancer Research Center, ACECR , Tehran , Iran
| | - Behrad Darvishi
- a Recombinant Proteins Department , Motamed Breast Cancer Research Center, ACECR , Tehran , Iran
| | - Keivan Majidzadeh-A
- b Genetics Department , Motamed Breast Cancer Research Center, ACECR , Tehran , Iran.,c Tasnim Biotechnology Research Center, Faculty of Medicine , AJA University of Medical Sciences , Tehran , Iran
| |
Collapse
|
27
|
Mei L, He Y, Wang H, Jin Y, Wang S, Jin C. Human hepatocyte growth factor inhibits early neointima formation in rabbit abdominal aortae following ultrasound-guided balloon injury. Mol Med Rep 2017; 16:5203-5210. [PMID: 28849185 PMCID: PMC5647058 DOI: 10.3892/mmr.2017.7229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 06/08/2017] [Indexed: 12/20/2022] Open
Abstract
The present study investigated the effects of in vivo gene transfer of human hepatocyte growth factor (hHGF) on neointima formation in rabbit abdominal aortae following ultrasound‑guided balloon injury. New Zealand white rabbits were randomly divided into four groups: endothelium injury alone (EI), endothelium injury with control vector transfection (EI‑V), endothelium injury with hHGF transfection (EI‑HGF), and hHGF transfection alone without endothelium injury (HGF). Endothelial injury was established by scraping the abdominal aortic wall using a balloon catheter under the guidance of a transabdominal ultrasound. hHGF gene transfer was performed 7 days following injury. hHGF mRNA and protein expression levels were determined at 3, 7, 14 and 21 days following transfection. Neointima formation was assessed by histopathological analysis at 14 and 28 days following injury. hHGF mRNA and protein expression levels were detected in the target abdominal aortae in EI‑HGF and HGF groups with the greatest levels observed 3 days following transfection, and their levels dropped below detection limits at 21 days following transfection. hHGF was not detectable in the EI and EI‑V groups throughout the experiment. The neointimal area and the neointima to media ratio in the EI‑HGF group were significantly decreased compared with those in the EI or EI‑V group at 14 days following injury. However, no differences were observed at 28 days following injury. The present study demonstrated that in vivo hHGF gene transfer inhibits the early formation of neointima in balloon‑injured rabbit abdominal aortae.
Collapse
Affiliation(s)
- Li Mei
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
- Department of Ultrasound, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yu He
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hui Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ying Jin
- Department of Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shuai Wang
- Department of Pathology, Cancer Hospital of Jilin, Changchun, Jilin 130012, P.R. China
| | - Chunxiang Jin
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
- Correspondence to: Professor Chunxiang Jin, Department of Ultrasound, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, Jilin 130033, P.R. China, E-mail:
| |
Collapse
|
28
|
Dahan N, Sarig U, Bronshtein T, Baruch L, Karram T, Hoffman A, Machluf M. Dynamic Autologous Reendothelialization of Small-Caliber Arterial Extracellular Matrix: A Preclinical Large Animal Study. Tissue Eng Part A 2016; 23:69-79. [PMID: 27784199 PMCID: PMC5240014 DOI: 10.1089/ten.tea.2016.0126] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Effective cellularization is a key approach to prevent small-caliber (<4 mm) tissue-engineered vascular graft (TEVG) failure and maintain patency and contractility following implantation. To achieve this goal, however, improved biomimicking designs and/or relatively long production times (typically several months) are required. We previously reported on porcine carotid artery decellularization yielding biomechanically stable and cell supportive small-caliber (3–4 mm diameter, 5 cm long) arterial extracellular matrix (scaECM) vascular grafts. In this study, we aimed to study the scaECM graft patency in vivo and possibly improve that patency by graft pre-endothelialization with the recipient porcine autologous cells using our previously reported custom-designed dynamic perfusion bioreactor system. Decellularized scaECM vascular grafts were histologically characterized, their immunoreactivity studied in vitro, and their biocompatibility profile evaluated as a xenograft subcutaneous implantation in a mouse model. To study the scaECM cell support and remodeling ability, pig autologous endothelial and smooth muscle cells (SMCs) were seeded and dynamically cultivated within the scaECM lumen and externa/media, respectively. Finally, endothelialized-only scaECMs—hypothesized as a prerequisite for maintaining graft patency and controlling intimal hyperplasia—were transplanted as an interposition carotid artery graft in a porcine model. Graft patency was evaluated through angiography online and endpoint pathological assessment for up to 6 weeks. Our results demonstrate the scaECM-TEVG biocompatibility preserving a structurally and mechanically stable vascular wall not just following decellularization and recellularization but also after implantation. Using our dynamic perfusion bioreactor, we successfully demonstrated the ability of this TEVG to support in vitro recellularization and remodeling by primary autologous endothelial and SMCs, which were seeded on the lumen and the externa/media layers, respectively. Following transplantation, dynamically endothelialized scaECM-TEVGs remained patent for 6 weeks in a pig carotid interposition bypass model. When compared with nonrevitalized control grafts, reendothelialized grafts provided excellent antithrombogenic activity, inhibited intimal hyperplasia formation, and encouraged media wall infiltration and reorganization with recruited host SMCs. We thus demonstrate that readily available decellularized scaECM can be promptly revitalized with autologous cells in a 3-week period before implantation, indicating applicability as a future platform for vascular reconstructive procedures.
Collapse
Affiliation(s)
- Nitsan Dahan
- 1 Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa, Israel
| | - Udi Sarig
- 1 Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa, Israel .,2 School of Materials Science and Engineering, Nanyang Technological University (NTU) , Singapore, Singapore
| | - Tomer Bronshtein
- 1 Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa, Israel
| | - Limor Baruch
- 1 Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa, Israel
| | - Tony Karram
- 3 Department of Vascular Surgery and Transplantation, Rambam Health Care Campus, Technion-Israel Institute of Technology , Haifa, Israel
| | - Aaron Hoffman
- 3 Department of Vascular Surgery and Transplantation, Rambam Health Care Campus, Technion-Israel Institute of Technology , Haifa, Israel
| | - Marcelle Machluf
- 1 Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology , Haifa, Israel .,2 School of Materials Science and Engineering, Nanyang Technological University (NTU) , Singapore, Singapore
| |
Collapse
|
29
|
Abstract
Coronary artery disease remains a major problem for Western societies. The advent of percutaneous interventions, including stents has brought clinical care to a new level of efficacy, yet problems remain. Restenosis following stenting in human coronary arteries appears at last to be yielding to therapeutic strategies, especially drug eluting stents. Because therapeutic percutaneous coronary intervention is widely dominated by the intracoronary stent, restenosis therapies must include the stented coronary artery. Animal models and in particular the porcine coronary model seem to represent the human coronary artery reaction to stenting. It mimics several clinical conditions including thrombosis and neointimal formation. A key question in the era of intravascular technologies is how well this and other models can predict clinical events. This paper discusses the models and their application.
Collapse
|
30
|
Jun I, Chung YW, Park J, Han HS, Park J, Kim S, Lee H, Kim SH, Han JH, Kim H, Seok HK, Kim YC, Jeon H. Ultrathin Metal Films with Defined Topographical Structures as In Vitro Cell Culture Platforms for Unveiling Vascular Cell Behaviors. Adv Healthc Mater 2016; 5:2396-405. [PMID: 27390259 DOI: 10.1002/adhm.201600333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/16/2016] [Indexed: 12/24/2022]
Abstract
Implanted material surfaces make direct contact with body tissues to work on its own purpose. Therefore, studies of the surface properties of implantable materials that determine cell fate are very important for successful implantation. Although numerous studies have addressed the relationship between cells and material surfaces, nonmetallic surfaces and metallic surfaces likely produce different cellular responses because of their intrinsic differences in surface energy, roughness, and chemical composition. Moreover, given the nontransparent property of metal materials, which hampers the real-time imaging of cellular behavior, a detailed cellular-level analysis at the metal-tissue interface has not been performed. In this study, metal-based cell culture platforms (MCPs) with defined microscale topographical patterns are developed using a combination of photolithography and direct current magnetron sputtering techniques. The MCPs allow to observe vascular cells on metals in real time and identify the selective regulation of human aortic smooth muscle cells and Human umbilical vein endothelial cells (HUVECs) by metallic surface topography. Additionally, atomic force microscopy, contact angles, and energy-dispersive X-ray spectroscopy analyses show that the MCPs exhibit nearly identical chemical properties with their bulk counterparts, demonstrating that MCPs can be utilized as an in vitro cell culture platform system for understanding the cellular behavior on metal substrates.
Collapse
Affiliation(s)
- Indong Jun
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Yong-Woo Chung
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Jimin Park
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Hyung-Seop Han
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Jaeho Park
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Saeromi Kim
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Hyunjung Lee
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Sang Hoon Kim
- Materials Architecturing Research Center; Materials and Life Science Research Division; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
| | - Jun-Hyun Han
- Department of Nano Materials Engineering; Chungnam National University; Daejeon 305-764 Republic of Korea
| | - Hyunjung Kim
- Division of Nursing; Hallym University; Chuncheon 24252 Republic of Korea
| | - Hyun-Kwang Seok
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
- Department of Bio-medical Engineering; Korea University of Science and Technology; Daejeon 34113 Republic of Korea
| | - Yu-Chan Kim
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
- Department of Bio-medical Engineering; Korea University of Science and Technology; Daejeon 34113 Republic of Korea
| | - Hojeong Jeon
- Center for Biomaterials; Biomedical Research Institute; Korea Institute of Science and Technology; Seoul 02792 Republic of Korea
- Department of Bio-medical Engineering; Korea University of Science and Technology; Daejeon 34113 Republic of Korea
| |
Collapse
|
31
|
Carr G, Barrese V, Stott JB, Povstyan OV, Jepps TA, Figueiredo HB, Zheng D, Jamshidi Y, Greenwood IA. MicroRNA-153 targeting of KCNQ4 contributes to vascular dysfunction in hypertension. Cardiovasc Res 2016; 112:581-589. [PMID: 27389411 PMCID: PMC5079273 DOI: 10.1093/cvr/cvw177] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 07/01/2016] [Indexed: 11/30/2022] Open
Abstract
Aims Kv7.4, a voltage-dependent potassium channel expressed throughout the vasculature, controls arterial contraction and is compromised in hypertension by an unknown mechanism. MicroRNAs (miRs) are post-transcriptional regulators of protein production and are altered in disease states such as hypertension. We investigated whether miRs regulate Kv7.4 expression. Methods and results In renal and mesenteric arteries (MAs) of the spontaneously hypertensive rat (SHR), Kv7.4 protein decreased compared with the normotensive (NT) rat without a decrease in KCNQ4 mRNA, inferring that Kv7.4 abundance was determined by post-transcriptional regulation. In silico analysis of the 3′ UTR of KCNQ4 revealed seed sequences for miR26a, miR133a, miR200b, miR153, miR214, miR218, and let-7d with quantitative polymerase chain reaction showing miR153 increased in those arteries from SHRs that exhibited decreased Kv7.4 levels. Luciferase reporter assays indicated a direct targeting effect of miR153 on the 3′ UTR of KCNQ4. Introduction of high levels of miR153 to MAs increased vascular wall thickening and reduced Kv7.4 expression/Kv7 channel function compared with vessels receiving a non-targeting miR, providing a proof of concept of Kv7.4 regulation by miR153. Conclusion This study is the first to define a role for aberrant miR153 contributing to the hypertensive state through targeting of KCNQ4 in an animal model of hypertension, raising the possibility of the use of miR153-related therapies in vascular disease.
Collapse
Affiliation(s)
- Georgina Carr
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Vincenzo Barrese
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Jennifer B Stott
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Oleksandr V Povstyan
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Thomas A Jepps
- Ion Channels Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hericka B Figueiredo
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Dongling Zheng
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Yalda Jamshidi
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Iain A Greenwood
- Vascular Research Centre, St George's University of London, Cranmer Terrace, London SW17 0RE, UK.,Ion Channels Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Prasad CK, Resmi KR, Krishnan LK, Vaishnav R. Survival of Endothelial Cells in vitro on Paclitaxel-loaded Coronary Stents. J Biomater Appl 2016; 19:271-86. [PMID: 15788425 DOI: 10.1177/0885328205047397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Coronary stents that are developed for use with balloon angioplasty are known to cause acute occlusion and long-term stenosis. It is likely that a controlled release of drugs at the site of stent implantation might inhibit the proliferation of vascular smooth muscle cells (VSMC) and reduce restenosis. However, if the drug is necrotic and affects cell survival near the implant, it may interrupt the local tissue regeneration. Different methods have been used for the immobilization of drugs with stents to get an effective concentration that inhibits cell proliferation. The objective of this study is to assess the effectiveness of Paclitaxel-loaded stents by immobilization with a biodegradable polymer, to inhibit cell proliferation. The cells used for the evaluation are human umbilical vein endothelial cells (HUVEC) and the proliferation rate of these cells on the drug-coated stent is compared against an uncoated stent for a 72-h period. Evaluations were also made to differentiate between cell apoptosis and necrosis to prove that the drug released is not deleterious to the surrounding tissue. While a similar initial cell adhesion is observed in bare and coated stents, the proliferation of HUVEC is negligible when grown on a drug-coated stent (p < 0.001). By specific staining techniques, the cells on the drug-coated stents are found to be apoptotic and not necrotic, throughout the evaluation period. In vitro leukocyte adhesion and platelet deposition on the drug-coated stents are found to be low when they are exposed to human blood and platelet-rich plasma (PRP), suggesting that the coated stents may not be thrombogenic in vivo. Therefore, drug coating of stents using the described technique may have a considerable promise for the prevention of neointimal proliferation, restenosis, and associated failure of angioplasty.
Collapse
Affiliation(s)
- C Krishna Prasad
- Thrombosis Research Unit, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Trivandrum-695 012, India
| | | | | | | |
Collapse
|
33
|
Wang P, Xu J, Hou Z, Wang F, Song Y, Wang J, Zhu H, Jin H. miRNA-34a promotes proliferation of human pulmonary artery smooth muscle cells by targeting PDGFRA. Cell Prolif 2016; 49:484-93. [PMID: 27302634 DOI: 10.1111/cpr.12265] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/07/2016] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Pulmonary arterial hypertension (PAH) is a fast progressing vascular disease characterized by uncontrolled cell proliferation of pulmonary artery smooth muscle cells (PASMCs). Some studies have suggested that PAH and cancers share an apoptosis-resistant state, featuring excessive cell proliferation. The miR-34 family consists of tumour-suppressive miRNAs, and its reduced expression has been reported in numerous cancers; however, its role in hypoxia-induced PAH has not been previously studied. MATERIALS AND METHODS miR-34 family expression was evaluated in a rat model with hypoxia and in cultured hypoxic PASMCs, using real-time quantitative PCR (RT-qPCR). Function of miR-34 family was assessed by transfecting miR-34 mimics and inhibitors. Dual luciferase reporter gene assays, RT-qPCR and Western blotting were performed to validate target genes of miR-34. RESULTS Significant down-regulation of miR-34a in hypoxic lung tissue, pulmonary artery and PASMCs was identified and then effects of miR-34a in modulating cell proliferation in human pulmonary artery smooth muscle cells (hPASMCs) was investigated in vitro. Reduction of miR-34a levels in hPASMCs caused increased proliferation and these effects were reversed by overexpression of miR-34a. miR-34a overexpression down-regulated platelet-derived growth factor receptor alpha (PDGFRA) expression, which is a key factor in PAH development. These results suggest that miR-34a is a potential regulator of proliferation in PASMCs, and that it could be used as a novel treatment strategy in PAH.
Collapse
Affiliation(s)
- Peng Wang
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China.,Department of Physiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, China
| | - Jie Xu
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Zhiling Hou
- Department of Emergency Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Fangfang Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jiamusi University, Jiamusi, China
| | - Yingli Song
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jiao Wang
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hui Zhu
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hongbo Jin
- Department of Physiology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Orynbayeva Z, Sensenig R, Polyak B. Metabolic and structural integrity of magnetic nanoparticle-loaded primary endothelial cells for targeted cell therapy. Nanomedicine (Lond) 2016; 10:1555-68. [PMID: 26008193 DOI: 10.2217/nnm.15.14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIM To successfully translate magnetically mediated cell targeting from bench to bedside, there is a need to systematically assess the potential adverse effects of magnetic nanoparticles (MNPs) interacting with 'therapeutic' cells. Here, we examined in detail the effects of internalized polymeric MNPs on primary rat endothelial cells' structural intactness, metabolic integrity and proliferation potential. MATERIALS & METHODS The intactness of cytoskeleton and organelles was studied by fluorescent confocal microscopy, flow cytometry and high-resolution respirometry. RESULTS MNP-loaded primary endothelial cells preserve intact cytoskeleton and organelles, maintain normal rate of proliferation, calcium signaling and mitochondria energy metabolism. CONCLUSION This study provides supportive evidence that MNPs at doses necessary for targeting did not induce significant adverse effects on structural integrity and functionality of primary endothelial cells - potential cell therapy vectors.
Collapse
Affiliation(s)
- Zulfiya Orynbayeva
- 1Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Richard Sensenig
- 2Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Boris Polyak
- 1Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| |
Collapse
|
35
|
Moyamoya Disease. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00040-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
Bass HM, Beard RS, Cha BJ, Yuan SY, Nelson PR. Thrombomodulin Induces a Quiescent Phenotype and Inhibits Migration in Vascular Smooth Muscle Cells In Vitro. Ann Vasc Surg 2015; 30:149-56. [PMID: 26549810 DOI: 10.1016/j.avsg.2015.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/14/2015] [Accepted: 10/27/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Loss of critical endothelial cell function and subsequent vascular smooth muscle cell (VSMC) migration is central to the pathology of injury-induced neointimal hyperplasia and recurrent stenosis. Thrombomodulin (TM), well known for its function as an endothelial surface anticoagulant, may have an unknown direct effect on VSMC physiology that would be lost after injury. Here, we examined a novel effect of TM on VSMC by testing the hypothesis that direct application of TM induces favorable changes to the morphology of VSMC and inhibits their migration. METHODS Primary human VSMC were harvested using the explant technique and used in early passage (1-4) for all experiments. Laser-scanning confocal fluorescent imaging was performed to assess the effect of soluble TM on VSMC morphology. In vitro, migration of VSMC was measured using: (1) a 4-hr modified Boyden chemotaxis assay and (2) a 24-hr electric cell-substrate impedance sensing injury migration assay. Migration experiments were conducted with VSMC exposed to increasing doses of soluble recombinant TM. Recombinant thrombin served as a positive control and serum-free media as a negative control for all experimentation. Data were analyzed using a Student's t-test or repeated measures analysis of variance where appropriate (α < 0.05). RESULTS VSMC exposed to TM clearly demonstrated a quiescent morphology with organized stress fibers consistent with a quiescent, differentiated, contractile phenotype; whereas, thrombin stimulation led to an activated, dedifferentiated, synthetic phenotype. VSMC demonstrated a low, baseline level of migration in unstimulated serum-free conditions. Thrombin significantly stimulated VSMC migration as expected. TM, independent of thrombin, significantly inhibited baseline VSMC migration in a dose-response fashion. The maximal inhibition was observed at (5 μg/mL) with 70% reduction (56 ± 1.7 vs. 18 ± 3.5 cells/5 high-power fields, P = 0.0005). CONCLUSIONS TM has a direct effect on VSMC resulting in a quiescent, differentiated and contractile phenotype, and inhibition of migration. This effect is independent of the presence of thrombin. These findings provide new knowledge in understanding the pathophysiology of vascular injury and support a strategy focused on restoring key endothelial function to prevent intimal hyperplasia.
Collapse
Affiliation(s)
- Heather M Bass
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Richard S Beard
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Byeong J Cha
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Peter R Nelson
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL; Division of Vascular and Cardiothoracic Surgery, Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL.
| |
Collapse
|
37
|
Ammann KR, DeCook KJ, Tran PL, Merkle VM, Wong PK, Slepian MJ. Collective cell migration of smooth muscle and endothelial cells: impact of injury versus non-injury stimuli. J Biol Eng 2015; 9:19. [PMID: 26473009 PMCID: PMC4606904 DOI: 10.1186/s13036-015-0015-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/15/2015] [Indexed: 01/19/2023] Open
Abstract
Background Cell migration is a vital process for growth and repair. In vitro migration assays, utilized to study cell migration, often rely on physical scraping of a cell monolayer to induce cell migration. The physical act of scrape injury results in numerous factors stimulating cell migration – some injury-related, some solely due to gap creation and loss of contact inhibition. Eliminating the effects of cell injury would be useful to examine the relative contribution of injury versus other mechanisms to cell migration. Cell exclusion assays can tease out the effects of injury and have become a new avenue for migration studies. Here, we developed two simple non-injury techniques for cell exclusion: 1) a Pyrex® cylinder - for outward migration of cells and 2) a polydimethylsiloxane (PDMS) insert - for inward migration of cells. Utilizing these assays smooth muscle cells (SMCs) and human umbilical vein endothelial cells (HUVECs) migratory behavior was studied on both polystyrene and gelatin-coated surfaces. Results Differences in migratory behavior could be detected for both smooth muscle cells (SMCs) and endothelial cells (ECs) when utilizing injury versus non-injury assays. SMCs migrated faster than HUVECs when stimulated by injury in the scrape wound assay, with rates of 1.26 % per hour and 1.59 % per hour on polystyrene and gelatin surfaces, respectively. The fastest overall migration took place with HUVECs on a gelatin-coated surface, with the in-growth assay, at a rate of 2.05 % per hour. The slowest migration occurred with the same conditions but on a polystyrene surface at a rate of 0.33 % per hour. Conclusion For SMCs, injury is a dominating factor in migration when compared to the two cell exclusion assays, regardless of the surface tested: polystyrene or gelatin. In contrast, the migrating surface, namely gelatin, was a dominating factor for HUVEC migration, providing an increase in cell migration over the polystyrene surface. Overall, the cell exclusion assays - the in-growth and out-growth assays, provide a means to determine pure migratory behavior of cells in comparison to migration confounded by cell wounding and injury. Electronic supplementary material The online version of this article (doi:10.1186/s13036-015-0015-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaitlyn R Ammann
- Biomedical Engineering GIDP, University of Arizona, Tucson, AZ USA
| | - Katrina J DeCook
- Biomedical Engineering GIDP, University of Arizona, Tucson, AZ USA
| | - Phat L Tran
- Sarver Heart Center, College of Medicine, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724 USA
| | - Valerie M Merkle
- Biomedical Engineering GIDP, University of Arizona, Tucson, AZ USA
| | - Pak K Wong
- Biomedical Engineering GIDP, University of Arizona, Tucson, AZ USA ; Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ USA
| | - Marvin J Slepian
- Biomedical Engineering GIDP, University of Arizona, Tucson, AZ USA ; Sarver Heart Center, College of Medicine, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724 USA ; Department of Biomedical Engineering, University of Arizona, Tucson, AZ USA
| |
Collapse
|
38
|
Chan JW, Zhang Y, Uhrich KE. Amphiphilic Macromolecule Self-Assembled Monolayers Suppress Smooth Muscle Cell Proliferation. Bioconjug Chem 2015; 26:1359-69. [PMID: 26042535 DOI: 10.1021/acs.bioconjchem.5b00208] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A significant limitation of cardiovascular stents is restenosis, where excessive smooth muscle cell (SMC) proliferation following stent implantation causes blood vessel reocclusion. While drug-eluting stents minimize SMC proliferation through releasing cytotoxic or immunosuppressive drugs from polymer carriers, significant issues remain with delayed healing, inflammation, and hypersensitivity reactions associated with drug and polymer coatings. Amphiphilic macromolecules (AMs) comprising a sugar-based hydrophobic domain and a hydrophilic poly(ethylene glycol) tail are noncytotoxic and recently demonstrated a concentration-dependent ability to suppress SMC proliferation. In this study, we designed a series of AMs and studied their coating properties (chemical composition, thickness, grafting density, and coating uniformity) to determine the effect of headgroup chemistry on bioactive AM grafting and release properties from stainless steel substrates. One carboxyl-terminated AM (1cM) and two phosphonate- (Me-1pM and Pr-1pM) terminated AMs, with varying linker lengths preceding the hydrophobic domain, were grafted to stainless steel substrates using the tethering by aggregation and growth (T-BAG) approach. The AMs formed headgroup-dependent, yet uniform, biocompatible adlayers. Pr-1pM and 1cM demonstrated higher grafting density and an extended release from the substrate over 21 days compared to Me-1pM, which exhibited lower grafting density and complete release within 7 days. Coinciding with their release profiles, Me-1pM and 1cM coatings initially suppressed SMC proliferation in vitro, but their efficacy decreased within 7 and 14 days, respectively, while Pr-1pM coatings suppressed SMC proliferation over 21 days. Thus, AMs with phosphonate headgroups and propyl linkers are capable of sustained release from the substrate and have the ability to suppress SMC proliferation during the restenosis that occurs in the 3-4 weeks after stent implantation, demonstrating the potential for AM coatings to provide sustained delivery via desorption from coated coronary stents and other metal-based implants.
Collapse
Affiliation(s)
- Jennifer W Chan
- †Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, United States
| | - Yingyue Zhang
- ‡Department of Chemistry and Chemical Biology, Rutgers University, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| | - Kathryn E Uhrich
- †Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, United States.,‡Department of Chemistry and Chemical Biology, Rutgers University, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| |
Collapse
|
39
|
Zhang J, Zhao F, Yu X, Lu X, Zheng G. MicroRNA-155 modulates the proliferation of vascular smooth muscle cells by targeting endothelial nitric oxide synthase. Int J Mol Med 2015; 35:1708-14. [PMID: 25872580 DOI: 10.3892/ijmm.2015.2181] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 03/26/2015] [Indexed: 11/05/2022] Open
Abstract
A variety of microRNAs (miRNAs) have been reported to be significantly be involved in the regulation of vascular smooth muscle cell (VSMC) proliferation, which is an essential process for the formation of atherosclerotic plaque. The objective of the present study was to explore the role of microRNA-155 (miR-155) in the regulation of VSMC growth and migration. A total of 12 atherosclerotic plaque samples and 9 control samples were collected, and the expression levels of miR-155/endothelial nitric oxide synthase (eNOS) were determined in those samples by RT-qPCR and western blot analysis. The results revealed that the relative expression levels of miR-155 in the atherosclerotic plaque samples were significantly upregulated compared with those in the normal control samples. We further found eNOS to be an effective target of miR-155 in the VSMCs by luciferase assay, which was confirmed by the observation that VSMCs transfected with miR-155 mimics exhibited a significantly lower protein expression level of eNOS. We also demonstrated that the exogenous overexpression of miR-155 significantly enhanced cell proliferation by inhibiting apoptosis in human aortic SMCs (HASMCs), and it also promoted the migratory ability of the cells. In conclusion, our data demonstrate that miR-155 is significantly upregulated in atherosclerotic plaque, functioning to accelerate the proliferation and migration of VSMCs by targeting eNOS.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Respiratory Disease, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110005, P.R. China
| | - Fei Zhao
- Department of Respiratory Disease, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110005, P.R. China
| | - Xiaoling Yu
- Department of Respiratory Disease, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110005, P.R. China
| | - Xiang Lu
- Department of Respiratory Disease, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110005, P.R. China
| | - Guofeng Zheng
- Department of Respiratory Disease, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110005, P.R. China
| |
Collapse
|
40
|
Cabiati M, Burchielli S, Matteucci M, Svezia B, Panchetti L, Caselli C, Prescimone T, Morales MA, Del Ry S. Dipyridamole-induced C-type natriuretic peptide mRNA overexpression in a minipig model of pacing-induced left ventricular dysfunction. Peptides 2015; 64:67-73. [PMID: 25613228 DOI: 10.1016/j.peptides.2015.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 12/23/2022]
Abstract
Dipyridamole (DP) restores ischemic tissue blood flow stimulating angiogenesis in eNOS-dependent pathways. C-type natriuretic peptide (CNP) is expected to mimic the migration-stimulatory effect of NO via a cGMP-dependent mechanism. Aim of this study was to assess the role of concomitant treatment with DP on CNP levels in blood and myocardial tissue of minipigs with left ventricular dysfunction (LVD) induced by pacing at 200bpm in the right ventricular apex. Minipigs with DP therapy (DP+, n=4) or placebo (DP-, n=4) and controls (C-SHAM, n=4) underwent 2D-EchoDoppler examination and blood collection before and after 4 weeks of pacing, when cardiac tissue was collected. Histological/immunohistochemical analyses were performed. CNP levels were determined by radioimmunoassay; cardiac CNP, BNP, natriuretic receptors expression by Real-Time PCR. After pacing, cardiac parameters resulted less impaired in DP+ compared to DP-. Histological sections presented normal morphology while the arteriolar density resulted: C-SHAM: 9.0±1.2; DP-: 4.9±0.3; DP+: 6.5±0.6number/mm(2); C-SHAM vs DP- and DP+ p=0.004, p=0.04, respectively. CNP mRNA resulted lower in DP- compared to C-SHAM and DP+ as well as NPR-B (p=0.011, DP- vs DP+). Both NPR-A/NPR-C mRNA expressions were significantly (p<0.001) lower both in DP- and DP+ compared to C-SHAM. BNP mRNA was higher in LVD. CNP plasma levels showed a similar trend with respect to gene expression (C-SHAM: 30.5±15; DP-: 18.6±5.5; DP+: 21.2±4.7pg/ml). These data suggest that DP may serve as a preconditioning agent to increase the protective CNP-mediated endocrine response in LVD. This response, mediated by its specific receptor NPR-B, may offer new insights into molecular targets for treatment of LVD.
Collapse
MESH Headings
- Animals
- Cardiac Pacing, Artificial
- Dipyridamole/pharmacology
- Dipyridamole/therapeutic use
- Disease Models, Animal
- Heart/drug effects
- Natriuretic Peptide, C-Type/genetics
- Natriuretic Peptide, C-Type/metabolism
- Protective Agents/pharmacology
- Protective Agents/therapeutic use
- RNA, Messenger/metabolism
- Swine
- Swine, Miniature
- Up-Regulation
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
Collapse
Affiliation(s)
- M Cabiati
- CNR Institute of Clinical Physiology, CNR, Italy
| | | | - M Matteucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - B Svezia
- CNR Institute of Clinical Physiology, CNR, Italy; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - L Panchetti
- Fondazione Toscana G. Monasterio, Pisa, Italy
| | - C Caselli
- CNR Institute of Clinical Physiology, CNR, Italy
| | - T Prescimone
- CNR Institute of Clinical Physiology, CNR, Italy
| | - M A Morales
- CNR Institute of Clinical Physiology, CNR, Italy
| | - S Del Ry
- CNR Institute of Clinical Physiology, CNR, Italy.
| |
Collapse
|
41
|
De Rosa S, Indolfi C. Circulating microRNAs as Biomarkers in Cardiovascular Diseases. EXPERIENTIA SUPPLEMENTUM (2012) 2015; 106:139-149. [PMID: 26608202 DOI: 10.1007/978-3-0348-0955-9_6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
MicroRNAs, key regulators of biological processes, are involved in the pathophysiological mechanisms underlying human diseases, including cardiovascular diseases. Their recent discovery revealed a previously unknown layer of pathophysiologic regulators, which also play a key role in the regulation of several aspects of cardiovascular diseases. More recently, it was demonstrated that circulating microRNAs can be measured in the blood. Hence, the potential use of microRNAs as disease biomarkers attracted many research groups. Indeed, their unusual stability in the bloodstream and during prolonged storage make circulating miRs very interesting as potential biomarkers. Circulating microRNAs are emerging as the next generation "smart" biomarkers and could be helpful in further improving the diagnostic and therapeutic processes of cardiovascular diseases. The present chapter summarizes the most relevant experimental evidence on circulating microRNAs in cardiovascular diseases, including arterial remodeling, restenosis, coronary artery disease, acute coronary syndromes, hypertension, heart failure, and ischemic stroke, highlighting potential pathophysiological correlations to the mechanisms underlying cardiovascular diseases.
Collapse
Affiliation(s)
- Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy. .,URT-CNR, Magna Graecia University, Catanzaro, 88100, Italy.
| |
Collapse
|
42
|
Mig-6 gene knockout induces neointimal hyperplasia in the vascular smooth muscle cell. DISEASE MARKERS 2014; 2014:549054. [PMID: 25574067 PMCID: PMC4276689 DOI: 10.1155/2014/549054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 11/27/2022]
Abstract
Although advances in vascular interventions can reduce the mortality associated with cardiovascular disease, neointimal hyperplasia remains a clinically significant obstacle limiting the success of current interventions. Identification of signaling pathways involved in migration and proliferation of vascular smooth muscle cells (SMCs) is an important approach for the development of modalities to combat this disease. Herein we investigate the role of an immediate early response gene, mitogen-inducible gene-6 (Mig-6), in the development of neointimal hyperplasia using vascular smooth muscle specific Mig-6 knockout mice. We induced endoluminal injury to one side of femoral artery by balloon dilatation in both Mig-6 knockout and control mice. Four weeks following injury, the artery of Mig-6 knockout mice demonstrated a 5.3-fold increase in the neointima/media ratio compared with control mice (P = 0.04). In addition, Mig-6 knockout vascular SMCs displayed an increase in both cell migration and proliferation compared with wild-type SMCs. Taken together, our data suggest that Mig-6 plays a critical role in the development of atherosclerosis. This finding provides new insight into the development of more effective ways to treat and prevent neointimal hyperplasia, particularly in-stent restenosis after percutaneous vascular intervention.
Collapse
|
43
|
Genetic risk of restenosis after percutaneous coronary interventions in the era of drug-eluting stents. Coron Artery Dis 2014; 25:658-64. [DOI: 10.1097/mca.0000000000000149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
44
|
Valoración de la lesión vascular tras la aplicación de métodos extraluminales de oclusión temporal. ANGIOLOGIA 2014. [DOI: 10.1016/j.angio.2014.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
45
|
Tolva V, Mazzola S, Zerbi P, Casana R, Albertini M, Calvillo L, Selmin F, Cilurzo F. A successful experimental model for intimal hyperplasia prevention using a resveratrol-delivering balloon. J Vasc Surg 2014; 63:788-94. [PMID: 25446284 DOI: 10.1016/j.jvs.2014.09.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/23/2014] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Restenosis due to intimal hyperplasia is a major clinical problem that compromises the success of angioplasty and endovascular surgery. Resveratrol (RSV) has demonstrated a beneficial effect on restenosis from angioplasty. Unfortunately, the physicochemical characteristics of RSV reduce the practicality of its immediate clinical application. This work proposes an experimental model aiming to setup an intravessel, elutable, RSV-containing compound. METHODS A 140 μg/mL RSV sterile injectable solution with a suitable viscosity for intravascular administration by drug-delivery catheter (RSV-c) was prepared. This solution was locally administered in the common iliac artery of adult male New Zealand White rabbits using a dedicated device (Genie; Acrostak, Geneva, Switzerland) after the induction of intimal hyperplasia by traumatic angioplasty. The RSV concentrations in the wall artery were determined, and the thickness of the harvested iliac arteries was measured over a 1-month period. RESULTS The Genie catheter was applied in rabbit vessels, and the local delivery resulted in an effective reduction in restenosis after plain angioplasty. Notably, RSV-c forced into the artery wall by balloon expansion might accumulate in the interstitial areas or within cells, avoiding the washout of solutions. Magnification micrographs showed intimal proliferation was significantly inhibited when RSV-c was applied. Moreover, no adverse events were documented in in vitro or in vivo studies. CONCLUSIONS RSV can be advantageously administered in the arterial walls by a drug-delivery catheter to reduce the risk of restenosis.
Collapse
Affiliation(s)
- Valerio Tolva
- Surgical Department, Istituto Auxologico Italiano Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Presidio Ospedaliero Capitanio, Milan, Italy; Dipartimento di Scienze della Salute, University of Milan-Bicocca, Milan, Italy.
| | - Silvia Mazzola
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi di Milano, Milan, Italy
| | - Pietro Zerbi
- Dipartimento di Scienze Biomediche e Cliniche "L. Sacco", Università degli Studi di Milano, Milan, Italy
| | - Renato Casana
- Surgical Department, Istituto Auxologico Italiano Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Presidio Ospedaliero Capitanio, Milan, Italy
| | - Mariangela Albertini
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi di Milano, Milan, Italy
| | - Laura Calvillo
- Department and Laboratory of Cardiac Arrhtmias on Genetic Basis, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Francesca Selmin
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milan, Italy
| | - Francesco Cilurzo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
46
|
Ren T, Yu S, Mao Z, Moya SE, Han L, Gao C. Complementary Density Gradient of Poly(hydroxyethyl methacrylate) and YIGSR Selectively Guides Migration of Endotheliocytes. Biomacromolecules 2014; 15:2256-64. [DOI: 10.1021/bm500385n] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tanchen Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Shan Yu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | | | - Lulu Han
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
47
|
Lee CH, Lin YH, Chang SH, Tai CD, Liu SJ, Chu Y, Wang CJ, Hsu MY, Chang H, Chang GJ, Hung KC, Hsieh MJ, Lin FC, Hsieh IC, Wen MS, Huang Y. Local sustained delivery of acetylsalicylic acid via hybrid stent with biodegradable nanofibers reduces adhesion of blood cells and promotes reendothelialization of the denuded artery. Int J Nanomedicine 2014; 9:311-26. [PMID: 24421640 PMCID: PMC3888352 DOI: 10.2147/ijn.s51258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Incomplete endothelialization, blood cell adhesion to vascular stents, and inflammation of arteries can result in acute stent thromboses. The systemic administration of acetylsalicylic acid decreases endothelial dysfunction, potentially reducing thrombus, enhancing vasodilatation, and inhibiting the progression of atherosclerosis; but, this is weakened by upper gastrointestinal bleeding. This study proposes a hybrid stent with biodegradable nanofibers, for the local, sustained delivery of acetylsalicylic acid to injured artery walls. Biodegradable nanofibers are prepared by first dissolving poly(D,L)-lactide-co-glycolide and acetylsalicylic acid in 1,1,1,3,3,3-hexafluoro-2-propanol. The solution is then electrospun into nanofibrous tubes, which are then mounted onto commercially available bare-metal stents. In vitro release rates of pharmaceuticals from nanofibers are characterized using an elution method, and a highperformance liquid chromatography assay. The experimental results suggest that biodegradable nanofibers release high concentrations of acetylsalicylic acid for three weeks. The in vivo efficacy of local delivery of acetylsalicylic acid in reducing platelet and monocyte adhesion, and the minimum tissue inflammatory reaction caused by the hybrid stents in treating denuded rabbit arteries, are documented. The proposed hybrid stent, with biodegradable acetylsalicylic acid-loaded nanofibers, substantially contributed to local, sustained delivery of drugs to promote re-endothelialization and reduce thrombogenicity in the injured artery. The stents may have potential applications in the local delivery of cardiovascular drugs. Furthermore, the use of hybrid stents with acetylsalicylic acid-loaded nanofibers that have high drug loadings may provide insight into the treatment of patients with high risk of acute stent thromboses.
Collapse
Affiliation(s)
- Cheng-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan ; Department of Mechanical Engineering, Taiwan
| | - Yu-Huang Lin
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taiwan
| | - Shang-Hung Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Chun-Der Tai
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taiwan
| | | | - Yen Chu
- Laboratory of Cardiovascular Physiology, Division of Thoracic and Cardiovascular Surgery, Taiwan
| | - Chao-Jan Wang
- Department of Medical Imaging and Intervention, Taiwan
| | - Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Taiwan
| | - Hung Chang
- Hematology-Oncology Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medicinal Sciences, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Kuo-Chun Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Ming-Jer Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Fen-Chiung Lin
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - I-Chang Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Yenlin Huang
- Department of Anatomical Pathology, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| |
Collapse
|
48
|
Affiliation(s)
- Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University
| | - Antonio Curcio
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University
- URT-CNR, Magna Graecia University
| |
Collapse
|
49
|
Muhammad R, Lim SH, Goh SH, Law JBK, Saifullah MSM, Ho GW, Yim EKF. Sub-100 nm patterning of TiO2 film for the regulation of endothelial and smooth muscle cell functions. Biomater Sci 2014; 2:1740-1749. [DOI: 10.1039/c4bm00212a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A sub-100 nm nano-imprinted TiO2 layer significantly inhibited the proliferation of SMCs and increased the proliferation of HUVECs. Focal adhesions size, density and distribution were significantly modulated by nano-imprinted TiO2.
Collapse
Affiliation(s)
- R. Muhammad
- Department of Biomedical Engineering
- National University of Singapore
- Singapore 117576, Republic of Singapore
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science
| | - S. H. Lim
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science
- Technology and Research)
- Singapore 117602, Republic of Singapore
- Department of Electrical and Computer Engineering
| | - S. H. Goh
- Department of Biomedical Engineering
- National University of Singapore
- Singapore 117576, Republic of Singapore
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science
| | - J. B. K. Law
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science
- Technology and Research)
- Singapore 117602, Republic of Singapore
| | - M. S. M. Saifullah
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science
- Technology and Research)
- Singapore 117602, Republic of Singapore
| | - G. W. Ho
- Department of Electrical and Computer Engineering
- National University of Singapore
- Singapore 117576, Republic of Singapore
| | - E. K. F. Yim
- Department of Biomedical Engineering
- National University of Singapore
- Singapore 117576, Republic of Singapore
- Mechanobiology Institute
- National University of Singapore
| |
Collapse
|
50
|
White RD, Holdaway BB, Moody JD, Chang Y. Chronic Caffeine Administration Attenuates Vascular Injury-Induced Neointimal Hyperplasia in Rats. JOURNAL OF CAFFEINE RESEARCH 2013; 3:163-168. [PMID: 24761282 DOI: 10.1089/jcr.2013.0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Inflammation is considered to be a major initiator to angioplasty-induced vascular restenosis. Proinflammatory cytokines stimulate vascular smooth muscle cell (VSMC) migration and proliferation leading to neointimal hyperplasia. It has been reported that chronic caffeine use suppresses the production of proinflammatory cytokine TNF-α (tumor necrosis factor Alpha) and alters adenosine receptor expression in human neutrophils, indicating that caffeine may attenuate vascular injury-induced inflammation and subsequent neointimal hyperplasia. Our current study was designed to test the hypothesis that chronic caffeine treatment decreases vascular injury-induced neointimal hyperplasia by suppressing VSMC migration and proliferation. Methods and Results: The experiments were carried out using both in vivo (rat carotid artery injury model) and in vitro (VSMCs isolated from rat aorta) models. Male Sprague-Dawley rats that received chronic caffeine treatment (10 and 20 mg/kg per day, through oral gavage) showed a significant decrease in neointimal hyperplasia when compared to rats that received vehicle. To understand the underlying mechanisms, we tested if caffeine inhibits fetal bovine serum (FBS)-induced VSMC migration and proliferation. We found that caffeine substantially suppressed FBS-induced VSMC migration and proliferation. The attenuation of FBS-stimulated cell migration is dose dependent. Conclusion: Together, our results suggest that chronic treatment with high concentrations of caffeine attenuates vascular injury-induced neointimal hyperplasia by suppressing smooth muscle cell migration and proliferation in rats.
Collapse
Affiliation(s)
- Ryan D White
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri. ; University of Missouri School of Medicine , Columbia, Missouri
| | - Brett B Holdaway
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri
| | - Joshua D Moody
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri
| | - Yingzi Chang
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri
| |
Collapse
|