1
|
Kaleli Durman D, Civelek E, Alp Yildirim Fİ, Teskin Ö, Uydeş Doğan BS. Comparative Vascular Effects of Sirolimus and Everolimus on Isolated Human Saphenous Veins. Life (Basel) 2025; 15:553. [PMID: 40283108 PMCID: PMC12028532 DOI: 10.3390/life15040553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Drug-eluting stents, which release antiproliferative agents such as sirolimus and everolimus, were developed to reduce the risk of restenosis associated with bare-metal stents. However, despite their proven clinical efficacy, concerns remain regarding in-stent restenosis due to delayed endothelial healing and the risk of late thrombotic events. In this study, we aimed to determine the vascular effects of sirolimus and everolimus on isolated human saphenous vein (SV) samples obtained from patients undergoing coronary artery bypass surgery. SV rings were subjected to sirolimus and everolimus in acute and pretreatment conditions in vitro. Increasing concentrations of sirolimus (10-8-10-5 M), everolimus (10-8-10-5 M), and their vehicle were administered to SV rings precontracted with phenylephrine (Phe,10-6-5 × 10-6 M) to evaluate their direct vascular effects. Additionally, SV rings were incubated (16 h) either with sirolimus (10-5 M), everolimus (10-6 M), or the vehicle. Thereafter, the contractile responses to Phe (10-8-10-4 M), and the endothelium-dependent and endothelium-independent relaxant responses to acetylcholine (ACh, 10-8-10-4 M) and sodium nitroprusside (SNP,10-8-10-4 M) were determined, respectively. Our findings demonstrated that sirolimus and everolimus did not exert direct relaxant and modulatory effects on vascular function in isolated human SVs. Hence, the preservation of contractile and relaxant responses with sirolimus and everolimus may have clinical implications in the context of DES implantation.
Collapse
Affiliation(s)
- Deniz Kaleli Durman
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Türkiye
| | - Erkan Civelek
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Türkiye
| | | | - Önder Teskin
- Department of Cardiovascular Surgery, Faculty of Medicine, Biruni University, 34015 Istanbul, Türkiye
| | | |
Collapse
|
2
|
Peters AA, Kaur C, Cabe M, Langert KA, Maier K, Gahtan V. Simvastatin-Loaded Chitosan-Functionalized PLGA Nanoparticles: Characterization and Use in Intimal Hyperplasia Therapy. Pharmaceutics 2025; 17:391. [PMID: 40143054 PMCID: PMC11944580 DOI: 10.3390/pharmaceutics17030391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Statins have beneficial pleiotropic effects, including reducing intimal hyperplasia (IH), but off-target effects remain a concern. Here, we tested the hypothesis that chitosan-functionalized polymeric nanoparticles (NPs) loaded with simvastatin (SL-cNPs) would (1) readily associate with endothelial cells (ECs) and vascular smooth muscle cells (VSMCs); (2) affect EC and VSMC function; and (3) reduce IH compared to systemic simvastatin. Methods: Human aortic ECs and VSMCs were cultured with fluorescently labeled SL-cNPs. The association of SL-cNPs was assessed by immunostaining and flow cytometry. The effect of SL-cNPs, empty cNPs (E-cNPs), and free simvastatin on cells was determined using qRT-PCR for RhoA and RhoB. Carotid artery balloon-injured rats were treated intraoperatively with intraluminal saline, E-cNPs, low- or high-dose SL-cNPs, periadventitial high-dose SL-cNPs, or with pre- and post-operative oral simvastatin plus intraoperative intraluminal saline or low-dose SL-cNPs. Rats were euthanized (day 14) and IH was quantified. Results: SL-cNPs readily associated with ECs and VSMCs. Low- and high-dose SL-cNPs induced significant increases in EC and VSMC RhoA gene expression. High-dose SL-cNPs induced a significant increase in EC RhoB expression, while free simvastatin and low- and high-dose SL-cNPs significantly increased RhoB expression in VSMCs. In vivo, oral simvastatin plus intraluminal SL-cNPs significantly reduced IH compared to controls. Conclusions: cNPs can be used as a vehicle to locally deliver statins to vascular cells. However, other NP formulations may be preferential for IH reduction given only the combination of oral simvastatin and SL-cNPs effectively reduced IH.
Collapse
Affiliation(s)
- Ashley A. Peters
- Department of Surgery, Loyola University of Chicago, Maywood, IL 60153, USA; (A.A.P.)
- Research Service, Edward Hines Jr., VA Hospital, Hines, IL 60141, USA
| | - Chanpreet Kaur
- Research Service, Edward Hines Jr., VA Hospital, Hines, IL 60141, USA
- Department of Molecular Pharmacology and Neuroscience, Loyola University of Chicago, Maywood, IL 60153, USA
| | - Maleen Cabe
- Research Service, Edward Hines Jr., VA Hospital, Hines, IL 60141, USA
- Department of Molecular Pharmacology and Neuroscience, Loyola University of Chicago, Maywood, IL 60153, USA
| | - Kelly A. Langert
- Research Service, Edward Hines Jr., VA Hospital, Hines, IL 60141, USA
- Department of Molecular Pharmacology and Neuroscience, Loyola University of Chicago, Maywood, IL 60153, USA
| | - Kristopher Maier
- Department of Surgery, Loyola University of Chicago, Maywood, IL 60153, USA; (A.A.P.)
| | - Vivian Gahtan
- Department of Surgery, Loyola University of Chicago, Maywood, IL 60153, USA; (A.A.P.)
- Research Service, Edward Hines Jr., VA Hospital, Hines, IL 60141, USA
- Surgical Service, Edward Hines Jr., VA Hospital, Hines, IL 60141, USA
| |
Collapse
|
3
|
Tierney JW, Francisco RP, Yu F, Ma J, Cheung-Flynn J, Keech MC, D'Arcy R, Shah VM, Kittel AR, Chang DJ, McCune JT, Bezold MG, Aligwekwe AN, Cook RS, Beckman JA, Brophy CM, Duvall CL. Intravascular delivery of an MK2 inhibitory peptide to prevent restenosis after angioplasty. Biomaterials 2025; 313:122767. [PMID: 39216327 PMCID: PMC12023847 DOI: 10.1016/j.biomaterials.2024.122767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Peripheral artery disease is commonly treated with balloon angioplasty, a procedure involving minimally invasive, transluminal insertion of a catheter to the site of stenosis, where a balloon is inflated to open the blockage, restoring blood flow. However, peripheral angioplasty has a high rate of restenosis, limiting long-term patency. Therefore, angioplasty is sometimes paired with delivery of cytotoxic drugs like paclitaxel to reduce neointimal tissue formation. We pursue intravascular drug delivery strategies that target the underlying cause of restenosis - intimal hyperplasia resulting from stress-induced vascular smooth muscle cell switching from the healthy contractile into a pathological synthetic phenotype. We have established MAPKAP kinase 2 (MK2) as a driver of this phenotype switch and seek to establish convective and contact transfer (coated balloon) methods for MK2 inhibitory peptide delivery to sites of angioplasty. Using a flow loop bioreactor, we showed MK2 inhibition in ex vivo arteries suppresses smooth muscle cell phenotype switching while preserving vessel contractility. A rat carotid artery balloon injury model demonstrated inhibition of intimal hyperplasia following MK2i coated balloon treatment in vivo. These studies establish both convective and drug coated balloon strategies as promising approaches for intravascular delivery of MK2 inhibitory formulations to improve efficacy of balloon angioplasty.
Collapse
Affiliation(s)
- J William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - R Paolo Francisco
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Chemical Engineering, School of Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA
| | - Veeraj M Shah
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Devin J Chang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Adrian N Aligwekwe
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; North Carolina State University, Raleigh, NC, 27695, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joshua A Beckman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, Nashville, TN, 37212, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
4
|
Li KR, Rohrich RN, Lava CX, Gupta NJ, Hidalgo CM, Episalla NC, Akbari CM, Evans KK. A Combined "Vasculoplastic" Approach to the Vasculopathic Patient Undergoing Limb Salvage: Understanding the Role of Endovascular Revascularization for Lower Extremity Free Tissue Transfer. J Reconstr Microsurg 2024. [PMID: 39730119 DOI: 10.1055/a-2491-3381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
BACKGROUND Multidisciplinary care with vascular surgery and plastic surgery is essential for lower extremity free flap (LEFF) success in the chronic wound population with diabetes and peripheral vascular disease. There is a lack of understanding on performing targeted direct endovascular reperfusion on a vessel that will be used as the flap recipient. Our study compares outcomes of patients who received targeted revascularization (TR) to the recipient vessel for LEFF anastomosis versus nontargeted revascularization (NR) of arterial recipients prior to LEFF. METHODS LEFF patients who underwent preoperative endovascular revascularization (ER) from July 2011 to January 2023 were reviewed. Location of ER, demographics, perioperative details, and outcomes were collected. TR was performed on the same vessel as the flap recipient and NR was located on a different vessel than the flap recipient. RESULTS A total of 55 LEFF patients were identified. Overall, 50.91% (n = 28) received TR and 49.1% (n = 27) received NR. Average age was 60.3 ± 10.9 years and average Charlson Comorbidity Index was 5.3 ± 1.9. On preoperative angiogram, the TR group had significantly lower rates of ER above the knee (3.6 vs. 33.3%, p < 0.001). Immediate flap success rate was 98.2%, with no differences between groups (p = 1.000). No significant differences were found in rates of any postoperative flap complications (p = 0.898), takeback (p = 0.352), partial flap necrosis (p = 0.648), or dehiscence (p = 0.729). Both TR and NR groups had similar rates of a postoperative angiogram (42.9 vs. 48.2%, p = 0.694) and reintervention (35.7 vs. 40.7% p = 0.701). Amputation rates were similar between TR and NR (17.9 vs. 14.8%, p = 1.000). CONCLUSION Close follow-up with vascular and plastic surgery is required for patients who undergo ER prior to LEFF, as nearly half of our cohort required additional endovascular procedures. Overall, we observed no significant differences in complication rates for the TR and NR groups, informing revascularization strategies for free tissue transfer in a highly comorbid chronic wound population.
Collapse
Affiliation(s)
- Karen R Li
- Department of Plastic and Reconstructive Surgery, MedStar Georgetown University Hospital, Washington, District of Columbia
- Georgetown University School of Medicine; Washington, District of Columbia
| | - Rachel N Rohrich
- Department of Plastic and Reconstructive Surgery, MedStar Georgetown University Hospital, Washington, District of Columbia
| | - Christian X Lava
- Department of Plastic and Reconstructive Surgery, MedStar Georgetown University Hospital, Washington, District of Columbia
- Georgetown University School of Medicine; Washington, District of Columbia
| | - Nisha J Gupta
- Georgetown University School of Medicine; Washington, District of Columbia
| | - Cecelia M Hidalgo
- Georgetown University School of Medicine; Washington, District of Columbia
| | - Nicole C Episalla
- Department of Plastic and Reconstructive Surgery, MedStar Georgetown University Hospital, Washington, District of Columbia
| | - Cameron M Akbari
- Department of Vascular Surgery, MedStar Georgetown University Hospital; Washington, District of Columbia
| | - Karen K Evans
- Department of Plastic and Reconstructive Surgery, MedStar Georgetown University Hospital, Washington, District of Columbia
| |
Collapse
|
5
|
Jadhav SA, Raval AJ, Jariwala AB, Engineer CB, Tailor J, Patravale VB. In vitro drug release profiling of Sirolimus polymeric microparticles coated long-acting stents. Int J Pharm 2024; 664:124572. [PMID: 39159857 DOI: 10.1016/j.ijpharm.2024.124572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024]
Abstract
In the realm of arterial disease interventions, drug-eluting stents (DES) have become a vital therapeutic choice in preventing atherosclerotic plaque formation and restenosis and facilitating vessel healing. Sirolimus-encapsulated poly Lactic-co-Glycolic acid (PLGA) Microparticles (MPs) were developed using solvent evaporation. MPs were freeze-dried with a cryoprotectant and coated on the stent surface using an efficient and reproducible nitrogen-assisted spray coating technique. The MPs displayed a uniform distribution particle size of 4.38 ± 1.1 μm, span value of 0.88 ± 0.02, coating mass transfer efficiency of 13.45 ± 1.1 % on the stent, and a coating time of ≤ 2 min per stent. Post sterilization, the particle size and morphology of the coated stents remained unchanged. Accelerated in vitro drug release profiles were evaluated under different conditions, indicating significant influences based on dissolution methods ranging from 28.2 %±4.3 %, 42.5 %±5.3 %, 76.6 %±4.7 %, and 84.25 %±3.1 % for dialysis bag (DB), vessel simulating flow-through cell (vFTC), flow-through cell (FTC), and sample and separate (SS) technique respectively for 48 h. The drug release mechanism from the coated stents is governed by the combination of the Korsmeyer Peppas and Higuchi models. The developed dissolution method exhibited discriminative effectiveness when evaluated with critical formulation attributes and process parameter variations. The 48 h accelerated drug release studies correlated well with the 6-month real-time release rate with an R2 value of 0.9142 and Pearson's R2 of 0.9561. Ex-vivo studies demonstrated the permeation of MPs into artery tissues. Stability studies confirmed that MPs coated stents maintained desired properties at 4 °C and 30 °C/65 % RH for 6 months. Overall, these findings contribute to advancing stent technology, suggesting the potential for improvement of arterial interventions and enhanced patient outcomes.
Collapse
Affiliation(s)
- Sarika A Jadhav
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga (E), Mumbai 400 019, Maharashtra, India
| | - Ankur J Raval
- Sahajanand Medical Technologies Limited, Gujarat 395004, India
| | | | | | - Jaynish Tailor
- Sahajanand Medical Technologies Limited, Gujarat 395004, India
| | - Vandana B Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga (E), Mumbai 400 019, Maharashtra, India.
| |
Collapse
|
6
|
Khoury M, Mekler T, Epshtein M, Kreinin Y, Korneyev D, Abezgauz L, Anagnostakou V, Ramon GZ, Sznitman J, Gounis M, Korin N. Isolation and focal treatment of brain aneurysms using interfacial fluid trapping. SCIENCE ADVANCES 2024; 10:eadp4579. [PMID: 39365869 PMCID: PMC11451524 DOI: 10.1126/sciadv.adp4579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/30/2024] [Indexed: 10/06/2024]
Abstract
Current approaches for localized intravascular treatments rely on using solid implants, such as metallic coils for embolizing aneurysms, or on direct injection of a therapeutic agent that can disperse from the required site of action. Here, we present a fluid-based strategy for localizing intravascular therapeutics that leverages surface tension and immiscible fluid interactions, to allow confined and focal treatment at brain aneurysm sites. We first show, computationally and experimentally, that an immiscible phase can be robustly positioned at the neck of human aneurysm models to seal and isolate the aneurysm's cavity for further treatment, including in wide-neck aneurysms. We then demonstrate localized delivery and confined treatment, by selective staining of cell nuclei within the aneurysm cavity as well as by hydrogel-based embolization in patient-specific aneurysm models. Altogether, our interfacial flow-driven strategy offers a potential approach for intravascular localized treatment of cardiovascular and other diseases.
Collapse
Affiliation(s)
- Maria Khoury
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
| | - Tirosh Mekler
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
| | - Mark Epshtein
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Yevgeniy Kreinin
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
| | - Dmitry Korneyev
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
| | - Ludmila Abezgauz
- Department of Environmental, Water and Agricultural Engineering, Technion - IIT, Haifa 32000, Israel
| | - Vania Anagnostakou
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Guy Z. Ramon
- Department of Environmental, Water and Agricultural Engineering, Technion - IIT, Haifa 32000, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
| | - Matthew Gounis
- Department of Radiology, New England Center for Stroke Research, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Netanel Korin
- Department of Biomedical Engineering, Technion - IIT, Haifa 32000, Israel
| |
Collapse
|
7
|
Braun SK, Jorge DW, Pedron VF. Influence of Preprocedural Statin Usage on Primary Patency and Amputation in Patients Undergoing Lower Limb Peripheral Angioplasty. Ann Vasc Surg 2024; 106:213-226. [PMID: 38821472 DOI: 10.1016/j.avsg.2024.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/24/2024] [Accepted: 03/24/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Peripheral arterial disease can progress to critical limb ischemia, which is associated with high amputation rates and requires revascularization. The endovascular approach has lower long-term patency because of restenosis due to neointimal hyperplasia. Statins are significantly advantageous for patients undergoing percutaneous interventions; however, only few studies have reported surgical improvements with statin therapy after endovascular treatment in such patients. This retrospective cohort study assessed the effects of preprocedural statins on lower limb arterial angioplasty outcomes by evaluating patency and amputation rates and comparing with those without statins. METHODS Patients who underwent percutaneous transluminal angioplasty of the lower limbs for critical ischemia of the lower limbs or for limiting claudication were included in this retrospective cohort study. Patients were categorized according to statin use prior to and during hospitalization. Patient demographics, lesion morphology, primary patency, and limb salvage rates were compared between these groups. Statistical analyses were performed using Kaplan-Meier and multivariate regression analysis. RESULTS A total of 178 patients undergoing endovascular intervention by critical ischemia or limiting claudication were included. Approximately 80% of the procedures were ballon angioplasty. Primary patency was 73% in 1 year and preprocedural statin usage was not associated with improved primary patency rates (P = 0.2798). After adjusting the amputation outcomes for pre-established variables, such as prehospitalization statin use, diabetes, procedure indication, disease location, Trans-Atlantic Inter-Society Consensus classification, and current smoking, there was no statistically significant difference associated with preprocedural statin use in primary patency (hazard ratio: 0.87 [0.33-2.29], P = 0.79) or amputation (hazard ratio: 0.70 [0.40-1.23], P = 0.22). CONCLUSIONS The use of preprocedural statin did not improve primary patency or amputation rates in patients undergoing peripheral angioplasty.
Collapse
Affiliation(s)
- Stela Karine Braun
- Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil; Surgery Department, Universidade Franciscana, Santa Maria, Rio Grande do Sul, Brazil.
| | | | | |
Collapse
|
8
|
Corti A, Marradi M, Çelikbudak Orhon C, Boccafoschi F, Büchler P, Rodriguez Matas JF, Chiastra C. Impact of Tissue Damage and Hemodynamics on Restenosis Following Percutaneous Transluminal Angioplasty: A Patient-Specific Multiscale Model. Ann Biomed Eng 2024; 52:2203-2220. [PMID: 38702558 PMCID: PMC11247064 DOI: 10.1007/s10439-024-03520-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024]
Abstract
Multiscale agent-based modeling frameworks have recently emerged as promising mechanobiological models to capture the interplay between biomechanical forces, cellular behavior, and molecular pathways underlying restenosis following percutaneous transluminal angioplasty (PTA). However, their applications are mainly limited to idealized scenarios. Herein, a multiscale agent-based modeling framework for investigating restenosis following PTA in a patient-specific superficial femoral artery (SFA) is proposed. The framework replicates the 2-month arterial wall remodeling in response to the PTA-induced injury and altered hemodynamics, by combining three modules: (i) the PTA module, consisting in a finite element structural mechanics simulation of PTA, featuring anisotropic hyperelastic material models coupled with a damage formulation for fibrous soft tissue and the element deletion strategy, providing the arterial wall damage and post-intervention configuration, (ii) the hemodynamics module, quantifying the post-intervention hemodynamics through computational fluid dynamics simulations, and (iii) the tissue remodeling module, based on an agent-based model of cellular dynamics. Two scenarios were explored, considering balloon expansion diameters of 5.2 and 6.2 mm. The framework captured PTA-induced arterial tissue lacerations and the post-PTA arterial wall remodeling. This remodeling process involved rapid cellular migration to the PTA-damaged regions, exacerbated cell proliferation and extracellular matrix production, resulting in lumen area reduction up to 1-month follow-up. After this initial reduction, the growth stabilized, due to the resolution of the inflammatory state and changes in hemodynamics. The similarity of the obtained results to clinical observations in treated SFAs suggests the potential of the framework for capturing patient-specific mechanobiological events occurring after PTA intervention.
Collapse
Affiliation(s)
- Anna Corti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, 20133, Milan, Italy.
| | - Matilde Marradi
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Cemre Çelikbudak Orhon
- Laboratory of Hemodynamics and Cardiovascular Technology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Philippe Büchler
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Jose F Rodriguez Matas
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Claudio Chiastra
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
9
|
Chung B, Wong M, Ch'ng JK. Toe Fillet Flap Wound Coverage for a Multiple-ray Amputation Wound: A Case Report. JOURNAL OF PLASTIC AND RECONSTRUCTIVE SURGERY 2024; 3:110-114. [PMID: 40103776 PMCID: PMC11913005 DOI: 10.53045/jprs.2023-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/20/2023] [Indexed: 03/20/2025]
Abstract
Ray amputation wounds caused by diabetic foot gangrene are often left to heal by secondary intention. They can be large and take a prolonged time to heal, exposing patients to complications and risk of recurrent infection. A 77-year-old male with diabetes and peripheral vascular disease presented to our institution with left 2nd-5th toe gangrene. He underwent a successful left lower limb angioplasty with good flow to the digital arteries. Left 2nd-5th toe ray amputation was performed, with the excess viable skin of the left second toe preserved as a digital fillet flap for wound coverage. The patient was discharged on postoperative day 1. Healing was complicated by a stitch sinus, but the wound completely healed with good epithelialization at 4 months postoperatively. This case report demonstrates the utility of the toe fillet flap in the coverage of ray amputation wounds in patients with diabetes and peripheral vascular disease.
Collapse
Affiliation(s)
- Benjamin Chung
- Department of Vascular Surgery, Singapore General Hospital, Singapore, Singapore
| | - Manzhi Wong
- Department of Plastic, Reconstructive and Aesthetic Surgery, Singapore General Hospital, Singapore, Singapore
| | - Jack Kian Ch'ng
- Department of Vascular Surgery, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
10
|
Chang TY, Lin MS, Chen CC, Leu YL, Wang SH. Isoxanthohumol reduces neointimal hyperplasia through the apelin/AKT pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167099. [PMID: 38428686 DOI: 10.1016/j.bbadis.2024.167099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
The abnormal proliferation, migration, and inflammation of vascular smooth muscle cells (VSMCs) play crucial roles in the development of neointimal hyperplasia and restenosis. Exposure to inflammatory cytokines such as platelet-derived growth factor (PDGF)-BB and tumour necrosis factor-alpha (TNF-α) induces the transformation of contractile VSMCs into abnormal synthetic VSMCs. Isoxanthohumol (IXN) has significant anti-inflammatory, antiproliferative, and antimigratory effects. This study aimed to explore the therapeutic impact and regulatory mechanism of IXN in treating neointimal hyperplasia. The present findings indicate that IXN effectively hinders the abnormal proliferation, migration, and inflammation of VSMCs triggered by PDGF or TNF-α. This inhibition is primarily achieved through the modulation of the apelin/AKT or AKT pathway, respectively. In an in vivo model, IXN effectively reduced neointimal hyperplasia in denuded femoral arteries. These results suggest that IXN holds promise as a potential and innovative therapeutic candidate for the treatment of restenosis.
Collapse
Affiliation(s)
- Ting-Yu Chang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mao-Shin Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chin-Chuan Chen
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan; Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan; Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
11
|
Fishbein I, Inamdar VV, Alferiev IS, Bratinov G, Zviman MM, Yekhilevsky A, Nagaswami C, Gardiner KL, Levy RJ, Stachelek SJ. Hypercholesterolemia exacerbates in-stent restenosis in rabbits: Studies of the mitigating effect of stent surface modification with a CD47-derived peptide. Atherosclerosis 2024; 390:117432. [PMID: 38241977 PMCID: PMC10939830 DOI: 10.1016/j.atherosclerosis.2023.117432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/07/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND AND AIMS Hypercholesterolemia (HC) has previously been shown to augment the restenotic response in animal models and humans. However, the mechanistic aspects of in-stent restenosis (ISR) on a hypercholesterolemic background, including potential augmentation of systemic and local inflammation precipitated by HC, are not completely understood. CD47 is a transmembrane protein known to abort crucial inflammatory pathways. Our studies have examined the interrelation between HC, inflammation, and ISR and investigated the therapeutic potential of stents coated with a CD47-derived peptide (pepCD47) in the hypercholesterolemic rabbit model. METHODS PepCD47 was immobilized on metal foils and stents using polybisphosphonate coordination chemistry and pyridyldithio/thiol conjugation. Cytokine expression in buffy coat-derived cells cultured over bare metal (BM) and pepCD47-derivatized foils demonstrated an M2/M1 macrophage shift with pepCD47 coating. HC and normocholesterolemic (NC) rabbit cohorts underwent bilateral implantation of BM and pepCD47 stents (HC) or BM stents only (NC) in the iliac location. RESULTS A 40 % inhibition of cell attachment to pepCD47-modified compared to BM surfaces was observed. HC increased neointimal growth at 4 weeks post BM stenting. These untoward outcomes were mitigated in hypercholesterolemic rabbits treated with pepCD47-derivatized stents. Compared to NC animals, inflammatory cytokine immunopositivity and macrophage infiltration of peri-strut areas increased in HC animals and were attenuated in HC rabbits treated with pepCD47 stents. CONCLUSIONS Augmented inflammatory responses underlie severe ISR morphology in hypercholesterolemic rabbits. Blockage of initial platelet and leukocyte attachment to stent struts through CD47 functionalization of stents mitigates the pro-restenotic effects of hypercholesterolemia.
Collapse
Affiliation(s)
- Ilia Fishbein
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| | - Vaishali V Inamdar
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ivan S Alferiev
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - George Bratinov
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Menekhem M Zviman
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | - Kristin L Gardiner
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Robert J Levy
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Stanley J Stachelek
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
12
|
Shazly T, Uline M, Webb C, Pederson B, Eberth JF, Kolachalama VB. Novel Payloads to Mitigate Maladaptive Inward Arterial Remodeling in Drug-Coated Balloon Therapy. J Biomech Eng 2023; 145:121004. [PMID: 37542712 PMCID: PMC10578076 DOI: 10.1115/1.4063122] [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] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
Drug-coated balloon therapy is a minimally invasive endovascular approach to treat obstructive arterial disease, with increasing utilization in the peripheral circulation due to improved outcomes as compared to alternative interventional modalities. Broader clinical use of drug-coated balloons is limited by an incomplete understanding of device- and patient-specific determinants of treatment efficacy, including late outcomes that are mediated by postinterventional maladaptive inward arterial remodeling. To address this knowledge gap, we propose a predictive mathematical model of pressure-mediated femoral artery remodeling following drug-coated balloon deployment, with account of drug-based modulation of resident vascular cell phenotype and common patient comorbidities, namely, hypertension and endothelial cell dysfunction. Our results elucidate how postinterventional arterial remodeling outcomes are altered by the delivery of a traditional anti-proliferative drug, as well as by codelivery with an anti-contractile drug. Our findings suggest that codelivery of anti-proliferative and anti-contractile drugs could improve patient outcomes following drug-coated balloon therapy, motivating further consideration of novel payloads in next-generation devices.
Collapse
Affiliation(s)
- Tarek Shazly
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208; College of Engineering and Computing, Department of Mechanical Engineering, University of South Carolina, Columbia, SC 29208; Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC 29208
| | - Mark Uline
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208; Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC 29208; College of Engineering and Computing, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208
| | - Clinton Webb
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208; Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC 29208; School of Medicine, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, SC 29208
| | - Breanna Pederson
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208
| | - John F. Eberth
- Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104
| | - Vijaya B. Kolachalama
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118; Department of Computer Science and Faculty of Computing and Data Sciences, Boston University, Boston, MA 02115
| |
Collapse
|
13
|
Cecchini AL, Biscetti F, Manzato M, Lo Sasso L, Rando MM, Nicolazzi MA, Rossini E, Eraso LH, Dimuzio PJ, Massetti M, Gasbarrini A, Flex A. Current Medical Therapy and Revascularization in Peripheral Artery Disease of the Lower Limbs: Impacts on Subclinical Chronic Inflammation. Int J Mol Sci 2023; 24:16099. [PMID: 38003290 PMCID: PMC10671371 DOI: 10.3390/ijms242216099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Peripheral artery disease (PAD), coronary artery disease (CAD), and cerebrovascular disease (CeVD) are characterized by atherosclerosis and inflammation as their underlying mechanisms. This paper aims to conduct a literature review on pharmacotherapy for PAD, specifically focusing on how different drug classes target pro-inflammatory pathways. The goal is to enhance the choice of therapeutic plans by considering their impact on the chronic subclinical inflammation that is associated with PAD development and progression. We conducted a comprehensive review of currently published original articles, narratives, systematic reviews, and meta-analyses. The aim was to explore the relationship between PAD and inflammation and evaluate the influence of current pharmacological and nonpharmacological interventions on the underlying chronic subclinical inflammation. Our findings indicate that the existing treatments have added anti-inflammatory properties that can potentially delay or prevent PAD progression and improve outcomes, independent of their effects on traditional risk factors. Although inflammation-targeted therapy in PAD shows promising potential, its benefits have not been definitively proven yet. However, it is crucial not to overlook the pleiotropic properties of the currently available treatments, as they may provide valuable insights for therapeutic strategies. Further studies focusing on the anti-inflammatory and immunomodulatory effects of these treatments could enhance our understanding of the mechanisms contributing to the residual risk in PAD and pave the way for the development of novel therapies.
Collapse
Affiliation(s)
- Andrea Leonardo Cecchini
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Federico Biscetti
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Matteo Manzato
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Lo Sasso
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Margherita Rando
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Anna Nicolazzi
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Enrica Rossini
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Luis H. Eraso
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paul J. Dimuzio
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Massimo Massetti
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Flex
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
14
|
Ji J, Xie Y, Bai Z, Deng G, Zhao G, Qin Y, Ji Z. Placement of Bare Self-Expanding Metal Stent for Isolated Superior Mesenteric Artery Dissection. Ann Vasc Surg 2023; 96:223-231. [PMID: 37230317 DOI: 10.1016/j.avsg.2023.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/20/2023] [Accepted: 04/30/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND To evaluate the safety and efficacy of placing bare self-expanding metal stent (SEMS) for treating isolated superior mesenteric artery dissection (ISMAD). METHOD Patients with ISMAD who received bare SEMS from January 2014 to December 2021 at the authors' center were included. Baseline characteristics, clinical manifestation, radiological findings, and treatment outcomes, including symptom relief and SMA remodeling, were analyzed. RESULT A total of 26 patients were included in this study. Among the patients, 25 were admitted due to persistent abdominal pain, and 1 was admitted based on computed tomography angiography (CTA) during physical examination. According to CTA scan, the percentage of stenosis was 91% (53.8-100%), and the length of dissection was 100.2 ± 8.4 mm. All patients received bare SEMS placement. The median time to symptom relief was 1 day (interquartile range, 1 3 days). The the median follow-up time of CTA was 6.8 months (range, 2-85 months), with an average of 16.2 months. Complete remodeling of the superior mesenteric artery (SMA) was recorded in 24 patients. The median time to remodeling was 3 months with an average of 4.7 months. Survival analysis indicated no significance difference in remodeling time between different ISMAD types based on Yun classification (P = 0.888) or between acute and nonacute disease (P = 0.423). Incomplete remodeling was noted in 2 patients. Distal stent occlusion without SMA-related symptoms was seen in 1 patient. Proximal stent stenosis occurred in 1 patient, and restenting was performed. The median follow-up time by telephone was 20.8 (4-91.5) months, and no intestinal ischemic symptoms were observed in any patient. CONCLUSIONS Bare SEMS placement can effectively relieve SMA-related symptoms in a short time and promote dissection remodeling in ISMAD. Time from symptom onset and classification of ISMAD seem not to have effects on SMA remodeling after bare SEMS placement.
Collapse
Affiliation(s)
- Jiajie Ji
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yiyun Xie
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Zhibin Bai
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Gang Deng
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Guofeng Zhao
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yonglin Qin
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| | - Zhenling Ji
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
15
|
Brodmann M, Wissgott C, Brechtel K, Lichtenberg M, Blessing E, Tarra T, Zeller T. Optimized drug-coated balloon angioplasty of the superficial femoral and proximal popliteal arteries using the Tack Endovascular System: Tack Optimized Balloon Angioplasty (TOBA) III 24-month results in standard and long lesions. Catheter Cardiovasc Interv 2023; 102:701-712. [PMID: 37560824 DOI: 10.1002/ccd.30800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/07/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE The Tack Endovascular System is a minimal-metal dissection repair device that is purpose-built to treat post-percutaneous angioplasty (PTA) arterial dissections in patients with peripheral arterial disease (PAD). The Tack Optimized Balloon Angioplasty (TOBA) III trial evaluated the safety and effectiveness of the Tack Endovascular System in patients with superficial femoral artery (SFA) and/or proximal popliteal artery (PPA) dissection after PTA with a drug-coated balloon (DCB). The objective of this study is to report the results in the standard- (SL) and long-lesion (LL) cohorts through 24 months. DESIGN The TOBA III study was a prospective, multicenter, single-arm study including patients suffering from Rutherford category 2-4 PAD. Outcomes were assessed according to pre-specified lesion length in SL ( ≥ 20 mm and ≤150 mm) and LL ( > 150 mm and ≤250 mm) cohorts. Follow-up was through 24 months. RESULTS TOBA III enrolled 201 patients, 169 patients in the SL cohort and 32 in the LL cohort. At 24 months, the Kaplan-Meier estimates of freedom from major adverse events were 91.7% and 82.6% for the SL cohort and LL cohort, respectively. Kaplan-Meier estimates of freedom from clinically driven-target lesion revascularization (CD-TLR) were 92.3% in the SL cohort and 82.6% in the LL cohort. At 24 months, 78.8% of SL patients and 69.2% of LL patients experienced an improvement of >2 Rutherford categories (both cohorts p < 0.001). The baseline ankle-brachial index improved from 0.68 ± 0.18 to 0.93 ± 0.16 in the SL (p < 0.001) and from 0.62 ± 0.23 to 0.87 ± 0.15 in the LL cohort (p < 0.001) at 24 months. CONCLUSION The 24-month results of the TOBA III trial support the safety and effectiveness of the Tack Endovascular System in patients who required post-PTA dissection repair in the SFA and PPA following DCB angioplasty for claudication and rest pain. In both the SL and LL cohorts, Tack placement was associated with sustained freedom from CD-TLR through 24 months as well as sustained improvements in Rutherford categories, ankle-brachial index, and quality of life.
Collapse
Affiliation(s)
| | | | - Klaus Brechtel
- Interventional Radiology, Franziskus-Hospital Berlin, Berlin, Germany
| | | | - Erwin Blessing
- SRH Klinikum Karlsbad-Langensteinbach, Karlsbad, Germany
| | - Trisha Tarra
- Philips North America, LLC, Cambridge, Massachusetts, USA
| | - Thomas Zeller
- Universitaets-Herzzentrum Freiburg - Bad Krozingen, Bad Krozingen, Germany
| |
Collapse
|
16
|
Ninno F, Tsui J, Balabani S, Díaz-Zuccarini V. A systematic review of clinical and biomechanical engineering perspectives on the prediction of restenosis in coronary and peripheral arteries. JVS Vasc Sci 2023; 4:100128. [PMID: 38023962 PMCID: PMC10663814 DOI: 10.1016/j.jvssci.2023.100128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/10/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Restenosis is a significant complication of revascularization treatments in coronary and peripheral arteries, sometimes necessitating repeated intervention. Establishing when restenosis will happen is extremely difficult due to the interplay of multiple variables and factors. Standard clinical and Doppler ultrasound scans surveillance follow-ups are the only tools clinicians can rely on to monitor intervention outcomes. However, implementing efficient surveillance programs is hindered by health care system limitations, patients' comorbidities, and compliance. Predictive models classifying patients according to their risk of developing restenosis over a specific period will allow the development of tailored surveillance, prevention programs, and efficient clinical workflows. This review aims to: (1) summarize the state-of-the-art in predictive models for restenosis in coronary and peripheral arteries; (2) compare their performance in terms of predictive power; and (3) provide an outlook for potentially improved predictive models. Methods We carried out a comprehensive literature review by accessing the PubMed/MEDLINE database according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The search strategy consisted of a combination of keywords and included studies focusing on predictive models of restenosis published between January 1993 and April 2023. One author independently screened titles and abstracts and checked for eligibility. The rest of the authors independently confirmed and discussed in case of any disagreement. The search of published literature identified 22 studies providing two perspectives-clinical and biomechanical engineering-on restenosis and comprising distinct methodologies, predictors, and study designs. We compared predictive models' performance on discrimination and calibration aspects. We reported the performance of models simulating reocclusion progression, evaluated by comparison with clinical images. Results Clinical perspective studies consider only routinely collected patient information as restenosis predictors. Our review reveals that clinical models adopting traditional statistics (n = 14) exhibit only modest predictive power. The latter improves when machine learning algorithms (n = 4) are employed. The logistic regression models of the biomechanical engineering perspective (n = 2) show enhanced predictive power when hemodynamic descriptors linked to restenosis are fused with a limited set of clinical risk factors. Biomechanical engineering studies simulating restenosis progression (n = 2) are able to capture its evolution but are computationally expensive and lack risk scoring for individual patients at specific follow-ups. Conclusions Restenosis predictive models, based solely on routine clinical risk factors and using classical statistics, inadequately predict the occurrence of restenosis. Risk stratification models with increased predictive power can be potentially built by adopting machine learning techniques and incorporating critical information regarding vessel hemodynamics arising from biomechanical engineering analyses.
Collapse
Affiliation(s)
- Federica Ninno
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Wellcome-EPSRC Centre for Interventional Surgical Sciences, London, United Kingdom
| | - Janice Tsui
- Department of Vascular Surgery, Royal Free Hospital NHS Foundation Trust, London, United Kingdom
- Division of Surgery & Interventional Science, Department of Surgical Biotechnology, Faculty of Medical Sciences, University College London, Royal Free Campus, London, United Kingdom
| | - Stavroula Balabani
- Wellcome-EPSRC Centre for Interventional Surgical Sciences, London, United Kingdom
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Vanessa Díaz-Zuccarini
- Wellcome-EPSRC Centre for Interventional Surgical Sciences, London, United Kingdom
- Department of Mechanical Engineering, University College London, London, United Kingdom
| |
Collapse
|
17
|
Shindo M, Hirotani S, Tsukada M, Morishita M. Venous Hypertension Improved by a Viabahn Stent Graft Blocking Regurgitation to the Periphery of the Basilic Vein in an Elderly Patient Undergoing Hemodialysis: A Case Report. J NIPPON MED SCH 2023; 90:351-355. [PMID: 35644560 DOI: 10.1272/jnms.jnms.2023_90-503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although necessary for hemodialysis (HD), arteriovenous grafts (AVG) frequently cause complications. Stenosis resulting in venous hypertension is a concern for physicians. Herein, we describe how venous hypertension was improved by using a Viabahn stent graft in an elderly HD patient. An 86-year-old woman started maintenance HD with a left-arm AVG. Two years later, she was referred to our hospital for treatment of juxta-graft-venous junction (GVJ) stenosis. Because of recurrence of stenosis at the juxta-GVJ, she underwent four percutaneous transluminal angioplasty (PTA) procedures during a period of 9 months. One month after the most recent PTA, the patient had redness, swelling, and pain in her left forearm. Venous hypertension was diagnosed on the basis of angiography findings showing regurgitation to the periphery of the basilic vein and juxta-GVJ stenosis. The stenosed juxta-GVJ was adequately expanded with a 7-mm balloon, and a 7-mm stent graft was inserted into the stenosis site. After successful treatment, there was no regurgitation to the periphery of the basilic vein and no symptoms. This complication should be considered when an AVG is created, because cutting off peripheral veins might prevent venous hypertension. Clinicians should perform regular postoperative monitoring.
Collapse
Affiliation(s)
- Mitsutoshi Shindo
- Department of Nephrology, Morishita Memorial Hospital
- Department of Dialysis Vascular Surgery, Morishita Memorial Hospital
| | - Sachiko Hirotani
- Department of Dialysis Vascular Surgery, Morishita Memorial Hospital
| | - Miho Tsukada
- Department of Nephrology, Morishita Memorial Hospital
| | - Masamitsu Morishita
- Department of Nephrology, Morishita Memorial Hospital
- Department of Dialysis Vascular Surgery, Morishita Memorial Hospital
| |
Collapse
|
18
|
Nardella E, Biscetti F, Rando MM, Cecchini AL, Nicolazzi MA, Rossini E, Angelini F, Iezzi R, Eraso LH, Dimuzio PJ, Pitocco D, Massetti M, Gasbarrini A, Flex A. Development of a biomarker panel for assessing cardiovascular risk in diabetic patients with chronic limb-threatening ischemia (CLTI): a prospective study. Cardiovasc Diabetol 2023; 22:136. [PMID: 37308885 DOI: 10.1186/s12933-023-01872-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Lower-extremity endovascular revascularization (LER) is often required for diabetic patients with chronic limb threatening ischemia (CLTI). During the post-revascularization period patients may unpredictably experience major adverse cardiac events (MACE) and major adverse limb events (MALE). Several families of cytokines are involved in the inflammatory process that underlies the progression of atherosclerosis. According to current evidence, we have identified a panel of possible biomarkers related with the risk of developing MACE and MALE after LER. The aim was to study the relationship between a panel of biomarkers - Interleukin-1 (IL-1) and 6 (IL-6), C-Reactive Protein (CRP), Tumor Necrosis Factor-α (TNF-α), High-Mobility Group Box-1 (HMGB-1), Osteoprotegerin (OPG), Sortilin and Omentin-1- at baseline, with cardiovascular outcomes (MACE and MALE) after LER in diabetic patients with CLTI. METHODS In this prospective non-randomized study, 264 diabetic patients with CLTI undergoing endovascular revascularization were enrolled. Serum levels of each biomarker were collected before revascularization and outcomes' incidence was evaluated after 1, 3, 6 and 12 months. RESULTS During the follow-up period, 42 cases of MACE and 81 cases of MALE occurred. There was a linear association for each biomarker at baseline and incident MACE and MALE, except Omentin-1 levels that were inversely related to the presence of MACE or MALE. After adjusting for traditional cardiovascular risk factors, the association between each biomarker baseline level and outcomes remained significant in multivariable analysis. Receiver operating characteristics (ROC) models were constructed using traditional clinical and laboratory risk factors and the inclusion of biomarkers significantly improved the prediction of incident events. CONCLUSIONS Elevated IL-1, IL-6, CRP, TNF-α, HMGB-1, OPG and Sortilin levels and low Omentin-1 levels at baseline correlate with worse vascular outcomes in diabetic patients with CLTI undergoing LER. Assessment of the inflammatory state with this panel of biomarkers may support physicians to identify a subset of patients more susceptible to the procedure failure and to develop cardiovascular adverse events after LER.
Collapse
Affiliation(s)
- Elisabetta Nardella
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
| | - Federico Biscetti
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy.
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy.
| | - Maria Margherita Rando
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
| | | | - Maria Anna Nicolazzi
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
| | - Enrica Rossini
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
| | - Flavia Angelini
- Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma, 00168, Italy
| | - Roberto Iezzi
- Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma, 00168, Italy
- Radiology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Luis H Eraso
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Paul J Dimuzio
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dario Pitocco
- Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma, 00168, Italy
- Diabetology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Massimo Massetti
- Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma, 00168, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
| | - Antonio Gasbarrini
- Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma, 00168, Italy
- Department of Medical and Surgical sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
| | - Andrea Flex
- Cardiovascular Internal Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, Roma, 00168, Italy
- Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma, 00168, Italy
| |
Collapse
|
19
|
Peng S, Wu WQ, Li LY, Shi YC, Lin S, Song ZY. Deficiency of neuropeptide Y attenuates neointima formation after vascular injury in mice. BMC Cardiovasc Disord 2023; 23:239. [PMID: 37149580 PMCID: PMC10164319 DOI: 10.1186/s12872-023-03267-y] [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: 10/08/2022] [Accepted: 04/26/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Restenosis after percutaneous coronary intervention (PCI) limits therapeutic revascularization. Neuropeptide Y (NPY), co-stored and co-released with the sympathetic nervous system, is involved in this process, but its exact role and underlying mechanisms remain to be fully understood. This study aimed to investigate the role of NPY in neointima formation after vascular injury. METHODS Using the left carotid arteries of wild-type (WT, NPY-intact) and NPY-deficient (NPY-/-) mice, ferric chloride-mediated carotid artery injury induced neointima formation. Three weeks after injury, the left injured carotid artery and contralateral uninjured carotid artery were collected for histological analysis and immunohistochemical staining. RT-qPCR was used to detect the mRNA expression of several key inflammatory markers and cell adhesion molecules in vascular samples. Raw264.7 cells were treated with NPY, lipopolysaccharide (LPS), and lipopolysaccharide-free, respectively, and RT-qPCR was used to detect the expression of these inflammatory mediators. RESULTS Compared with WT mice, NPY-/- mice had significantly reduced neointimal formation three weeks after injury. Mechanistically, immunohistochemical analysis showed there were fewer macrophages and more vascular smooth muscle cells in the neointima of NPY-/- mice. Moreover, the mRNA expression of key inflammatory markers such as interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), and intercellular adhesion molecule-1 (ICAM-1) was significantly lower in the injured carotid arteries of NPY-/- mice, compared to that in the injured carotid arteries of WT mice. In RAW264.7 macrophages, NPY significantly promoted TGF-β1 mRNA expression under unactivated but not LPS-stimulated condition. CONCLUSIONS Deletion of NPY attenuated neointima formation after artery injury, at least partly, through reducing the local inflammatory response, suggesting that NPY pathway may provide new insights into the mechanism of restenosis.
Collapse
Affiliation(s)
- Song Peng
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei-Qiang Wu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin-Yu Li
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan-Chuan Shi
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Zhi-Yuan Song
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
20
|
Zhao W, Chu X, Song Y, Zhang J, Sun L, Zheng M, Yin H, Zhang J, Wang W, Meng Y, Han J. Drug-Coated Balloon Treatment for Delayed Recanalization of Symptomatic Intracranial Artery Occlusion. Transl Stroke Res 2023; 14:193-199. [PMID: 35460456 PMCID: PMC9995415 DOI: 10.1007/s12975-022-01024-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/11/2022] [Accepted: 04/16/2022] [Indexed: 11/26/2022]
Abstract
Patients with medically refractory non-acute intracranial artery occlusion (ICAO) are difficult to treat. The optimal intervention for these patients is not known. We evaluated the feasibility and safety of drug-coated balloon (DCB) treatment for non-acute ICAO. Consecutive patients with symptomatic medically refractory atherosclerotic non-acute ICAO from January 2015 to July 2021 who underwent DCB treatment were retrospectively analyzed. The rates of stroke, transient ischemic attack, and death within 30 days and the follow-up results were evaluated. A total of 148 patients were enrolled in this study. The 30-day rate of stroke, transient ischemic attack, and death was 8.8%. During the 25.8 ± 15.8-month clinical follow-up period, the rate of outcome beyond 30 days was 4.7%. In the 66 patients with vessel imaging follow-up, 13.6% (9/66) had restenosis. The present study suggests that DCB dilatation is a feasible and effective alternative in carefully selected patients with symptomatic non-acute ICAO.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Xi Chu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Yun Song
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Jinping Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Lili Sun
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Meimei Zheng
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Hao Yin
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Jun Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Wei Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Yao Meng
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| | - Ju Han
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766#, Jingshi Road, Jinan, China
| |
Collapse
|
21
|
Okuno S, Iida O, Takahara M, Hata Y, Kurata N, Toyoshima T, Asai M, Masuda M, Okamoto S, Ishihara T, Nanto K, Kanda T, Tsujimura T, Matsuda Y, Mano T. Impact of intravascular ultrasound parameters and platelet reactivity on primary patency after drug-coated balloon angioplasty for femoropopliteal artery disease. Heart Vessels 2023; 38:497-506. [PMID: 36454300 DOI: 10.1007/s00380-022-02201-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/10/2022] [Indexed: 12/04/2022]
Abstract
Although the superiority of DCBs to uncoated balloon angioplasty for the treatment of femoropopliteal (FP) lesions has been demonstrated, the association of clinical factors, including anatomical features evaluated by intravascular ultrasound (IVUS) and platelet reactivity, with the loss of patency has not been systematically studied. The current prospective, observational study enrolled 160 consecutive patients (male 67.5%, mean age 74.7 ± 9.7 years) with 213 FP lesions treated with DCBs under IVUS evaluation. The platelet reactivity was measured in P2Y12 reaction units for all of the patients at the DCB treatment. The primary end point was primary patency at 12 months, while the secondary end points were freedom from target lesion revascularization (TLR), all-cause death, major target limb amputation and bleeding events at 12 months. Mean lesion length was 11.9 ± 9.4 cm and 34 (16.0%) were chronic total occlusions (CTOs). Thirty-four (16.0%) were severely calcified lesions. Primary patency by Kaplan-Meier estimate was 79.2% at 12 months, while the 12-month freedom from TLR, all-cause death and bleeding events were observed in 89.1%, 93.4% and 97.4%, respectively. There were no major target limb amputations through 12 months. Multivariate analysis showed that subintimal angioplasty for CTO lesions was a sole risk factor for loss of 12-month primary patency, while other IVUS parameters and platelet reactivity were not.
Collapse
Affiliation(s)
- Shota Okuno
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Osamu Iida
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan.
| | - Mitsuyoshi Takahara
- Department of Diabetes Care Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yosuke Hata
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Naoya Kurata
- Department of Clinical Engineering, Kansai Rosai Hospital, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Taku Toyoshima
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Mitsutoshi Asai
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Masaharu Masuda
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Shin Okamoto
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Takayuki Ishihara
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Kiyonori Nanto
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Takashi Kanda
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Takuya Tsujimura
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Yasuhiro Matsuda
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Toshiaki Mano
- Kansai Rosai Hospital Cardiovascular Center, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| |
Collapse
|
22
|
Subedi U, Manikandan S, Bhattarai S, Sharma P, Sharma S, Sun H, Miriyala S, Panchatcharam M. The Autotaxin-LPA Axis Emerges as a Novel Regulator of Smooth Muscle Cell Phenotypic Modulation during Intimal Hyperplasia. Int J Mol Sci 2023; 24:2913. [PMID: 36769255 PMCID: PMC9917461 DOI: 10.3390/ijms24032913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Neointimal hyperplasia is characterized by a loss of the contractile phenotype of vascular smooth muscle cells (VSMCs). Our group has recently shown that VSMC proliferation and migration are mediated by lysophosphatidic acid (LPA) during restenosis, but the role of autotaxin (ATX; lysophospholipase D), which produces LPA, remains unclear. Endothelial denudation of the mouse carotid artery was performed to induce neointimal hyperplasia, and the extent of damage caused by the ATX-LPA axis was assessed in VSMCs. We observed the upregulation of ATX activity (p < 0.0002) in the injured carotid artery using an AR2 probe fluorescence assay. Further, the tissue carotid LPA levels were elevated 2.7-fold in carotid vessels, augmenting neointimal hyperplasia. We used an electrical cell-substrate impedance sensor (ECIS) to measure VSMC proliferation and migration. Treatment with an ATX inhibitor (PF8380) or LPA receptor inhibitor (Ki16425) attenuated VSMC proliferation (extracellular signal-regulated kinases) activity and migration in response to recombinant ATX. Indeed, PF8380 treatment rescued the aggravated post-wire injury neointima formation of carotid arteries. The upregulation of ATX following vessel injury leads to LPA production in VSMCs, favoring restenosis. Our observations suggest that inhibition of the ATX-LPA axis could be therapeutically targeted in restenosis to minimize VSMC phenotypic modulation and inflammation after vascular injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences-Shreveport, Shreveport, LA 71130, USA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences-Shreveport, Shreveport, LA 71130, USA
| |
Collapse
|
23
|
Shazly T, Torres WM, Secemsky EA, Chitalia VC, Jaffer FA, Kolachalama VB. Understudied factors in drug-coated balloon design and evaluation: A biophysical perspective. Bioeng Transl Med 2023; 8:e10370. [PMID: 36684110 PMCID: PMC9842065 DOI: 10.1002/btm2.10370] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/28/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023] Open
Abstract
Drug-coated balloon (DCB) percutaneous interventional therapy allows for durable reopening of the narrowed lumen via physical tissue expansion and local anti-restenosis drug delivery, providing an alternative to traditional uncoated balloons or a permanent indwelling implant such as a conventional metallic drug-eluting stent. While DCB-based treatment of peripheral arterial disease (PAD) has been incorporated into clinical guidelines, DCB use has been recently curtailed due to reports that showed evidence of increased mortality risk in patients treated with paclitaxel (PTX)-coated balloons. Given the United States Food and Drug Administration's 2019 consequent warning regarding PTX-eluting DCBs and the subsequent marked reduction in clinical DCB use, there is now a critical need to better understand the compositional and mechanical factors underlying DCB efficacy and safety. Most work to date on DCB refinement has focused on designing both the enabling balloon catheter and alternate coatings composed of various drugs and excipients, followed by device evaluation in preclinical and clinical studies. We contend that improvement in DCB performance will require a better understanding of the biophysical factors operative during and following balloon deployment, and moreover that the elaboration and demonstrated control of these factors are needed to address current concerns with DCB use. This article provides a perspective on the biophysical interactions that govern DCB performance and offers new design strategies for the development of next-generation DCB devices.
Collapse
Affiliation(s)
- Tarek Shazly
- College of Engineering & ComputingUniversity of South CarolinaColumbiaSouth CarolinaUSA
| | - William M. Torres
- College of Engineering & ComputingUniversity of South CarolinaColumbiaSouth CarolinaUSA
- Exponent Inc.PhiladelphiaPennsylvaniaUSA
| | - Eric A. Secemsky
- Smith Center for Outcomes Research in CardiologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Vipul C. Chitalia
- Department of Medicine, Boston University School of MedicineBoston Veterans Affairs Healthcare SystemBostonMassachusettsUSA
| | - Farouc A. Jaffer
- Cardiovascular Research Center and Cardiology DivisionMassachusetts General HospitalBostonMassachusettsUSA
| | - Vijaya B. Kolachalama
- Department of Medicine, Boston University School of Medicine; Department of Computer Science and Faculty of Computing & Data SciencesBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
24
|
Multiscale agent-based modeling of restenosis after percutaneous transluminal angioplasty: Effects of tissue damage and hemodynamics on cellular activity. Comput Biol Med 2022; 147:105753. [DOI: 10.1016/j.compbiomed.2022.105753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
|
25
|
Tang TY, Sulaiman MSB, Soon SXY, Yap CJQ, Patel A, Chong TT. Slow-flow phenomena following lower limb paclitaxel- and sirolimus-coated balloon angioplasty in the setting of chronic limb threatening ischaemia-a case series. Quant Imaging Med Surg 2022; 12:2058-2065. [PMID: 35284292 PMCID: PMC8899964 DOI: 10.21037/qims-21-633] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/04/2021] [Indexed: 09/03/2024]
Abstract
The Achilles heel of plain old balloon angioplasty (POBA) is neointimal hyperplasia (NIH) and restenosis, caused from the barotrauma of ballooning. Drug-coated balloons using a paclitaxel-based platform (PCB) have been shown to retard the restenotic process, using the anti-proliferative effects of paclitaxel, and give longer vessel patency. This is important in the setting of chronic limb threatening ischemia (CLTI) and for the protracted wound healing process in these frail patients. However, during PCB application, more than 50% of the drug is lost downstream, a phenomenon termed particulate embolization. This is thought to account for the slow- or no-flow phenomenon encountered after PCB use. Recent data suggest that slow-flow phenomenon was associated with a lower target lesion revascularisation rate and worse amputation free survival (AFS). The use of sirolimus coated balloons (SCB) to impede the NIH cascade has been less well studied but recent data suggested excellent short-term efficacy and found no slow flow phenomenon with their use in the tibial arteries in CLTI patients. Aim of this case series is to highlight the difference in flow phenomena using PCB and SCB elution in the setting of CLTI. We evaluated the use of parametric colour coding and time attenuation curves (TAC) as a quantitative measure of blood flow. SCB may have an advantage over PCB use in the peripheral vasculature because of a reduced incidence of slow flow phenomenon following drug elution.
Collapse
Affiliation(s)
- Tjun Y. Tang
- Department of Vascular Surgery, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Shereen X. Y. Soon
- Department of Vascular Surgery, Singapore General Hospital, Singapore, Singapore
| | - Charyl J. Q. Yap
- Department of Vascular Surgery, Singapore General Hospital, Singapore, Singapore
| | - Ankur Patel
- Department of Vascular Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Tze T. Chong
- Department of Vascular Surgery, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
26
|
Fan XZ, Wang YY, Cui ZY, Cheng ZH, Zhang HL, Gamper N, Zhang F, Han M. Kv7.4 channel is a key regulator of vascular inflammation and remodeling in neointimal hyperplasia and abdominal aortic aneurysms. Free Radic Biol Med 2022; 178:111-124. [PMID: 34863875 DOI: 10.1016/j.freeradbiomed.2021.11.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
Inflammation has recently emerged as an important contributor for cardiovascular disease development and participates pivotally in the development of neointimal hyperplasia and abdominal aortic aneurysms (AAA) formation. Kv7.4/KCNQ4, a K+ channel, is one of the important regulators of vascular function but its role in vascular inflammation is unexplored. Here, we showed that the expression of Kv7.4 channel was elevated in the neointima and AAA tissues from mice and humans. Genetic deletion or pharmacological inhibition of Kv7.4 channel in mice alleviated neointimal hyperplasia and AAA formation via downregulation of a set of vascular inflammation-related genes, matrix metalloproteinases (MMP) 2/9, and intercellular adhesion molecule (ICAM-1). Furthermore, genetic deletion or inhibition of Kv7.4 channel suppressed the activation of tumor necrosis factor receptor 1 (TNFR1)-nuclear factor (NF)-κB signaling pathway via blockade of interaction between TNFR1 and TNFR1-associated death domain protein (TRADD) in vascular smooth muscle cells (VSMCs). Knockdown of Kv7.4 in vivo identified VSMC-expressed Kv7.4 as a major factor in vascular inflammation. Collectively, our findings suggest that Kv7.4 channel aggravates vascular inflammatory response, which promotes the neointimal hyperplasia and AAA formation. Inhibition of Kv7.4 channel may be a novel therapeutic strategy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- Xi-Zhenzi Fan
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Ying-Ying Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Hao Cheng
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Hai-Lin Zhang
- Department of Pharmacology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, PR China; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Nikita Gamper
- Department of Pharmacology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, PR China; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Fan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
27
|
Biscetti F, Cecchini AL, Rando MM, Nardella E, Gasbarrini A, Massetti M, Flex A. Principal predictors of major adverse limb events in diabetic peripheral artery disease: A narrative review. ATHEROSCLEROSIS PLUS 2021; 46:1-14. [PMID: 36643723 PMCID: PMC9833249 DOI: 10.1016/j.athplu.2021.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/10/2021] [Accepted: 10/28/2021] [Indexed: 01/18/2023]
Abstract
Background and aims The increasing prevalence of diabetes mellitus is causing a massive growth of peripheral artery disease incidences, a disabling complication of diabetic atherosclerosis, which leads often to the amputation of the affected limb. Critical limb ischemia is the terminal disease stage, which requires a prompt intervention to relieve pain and save limbs. However, patients undergoing revascularization often suffer from cardiovascular, cerebrovascular and major adverse limb events with poor outcomes. Furthermore, the same procedure performed in apparently similar patients has various outcomes and lack of an outcome predictive support causes a high lower limb arterial revascularization rate with disastrous effects for patients. We collected the main risk factors of major adverse limb events in a more readable and immediate format of the topic, to propose an overview of parameters to manage effectively peripheral artery disease patients and to propose basics of a new predictive tool to prevent from disabling vascular complications of the disease. Methods Most recent and updated literature about the prevalence of major adverse limb events in peripheral artery disease was reviewed to identify possible main predictors. Results In this article, we summarized major risk factors of limb revascularization failure and disabling vascular complications collecting those parameters principally responsible for major adverse limb events, which provides physio-pathological explanation of their role in peripheral artery disease. Conclusion We evaluated and listed a panel of possible predictors of MALE (Major Adverse Limb Event) in order to contribute to the development of a predictive score, based on a summary of the main risk factors reported in scientific articles, which could improve the management of peripheral artery disease by preventing vascular accidents.
Collapse
Affiliation(s)
- Federico Biscetti
- Internal and Cardiovascular Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy,Department of Cardiovascular Sciences, Università Cattolica del Sacro Cuore, Roma, Italy,Corresponding author. Internal and Cardiovascular Medicine Unit. Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | | | - Maria Margherita Rando
- Department of Cardiovascular Sciences, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Elisabetta Nardella
- Department of Cardiovascular Sciences, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Universitá Cattolica del Sacro Cuore, Roma, Italy
| | - Massimo Massetti
- Department of Cardiovascular Sciences, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Andrea Flex
- Internal and Cardiovascular Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy,Department of Medical and Surgical Sciences, Universitá Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
28
|
Wu J, Jin L, Tan JY, Chen XF, Wang QQ, Yuan GY, Chen TX. The effects of a biodegradable Mg-based alloy on the function of VSMCs via immunoregulation of macrophages through Mg-induced responses. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1292. [PMID: 34532429 PMCID: PMC8422083 DOI: 10.21037/atm-21-1375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Background Restenosis is one of the worst side effects of percutaneous coronary intervention (PCI) due to neointima formation resulting from the excessive proliferation and migration of vascular smooth muscle cells (VSMCs) and continuous inflammation. Biodegradable Mg-based alloy is a promising candidate material because of its good mechanical properties and biocompatibility, and biodegradation of cardiovascular stents. Although studies have shown reduced neointima formation after Mg-based CVS implantation in vivo, these findings were inconsistent with in vitro studies, demonstrating magnesium-mediated promotion of the proliferation and migration of VSMCs. Given the vital role of activated macrophage-driven inflammation in neointima formation, along with the well-demonstrated crosstalk between macrophages and VSMCs, we investigated the interactions of a biodegradable Mg-Nd-Zn-Zr alloy (denoted JDBM), which is especially important for cardiovascular stents, with VSMCs via macrophages. Methods JDBM extracts and MgCl2 solutions were prepared to study their effect on macrophages. To study the effects of the JDBM extracts and MgCl2 solutions on the function of VSMCs via immunoregulation of macrophages, conditioned media (CM) obtained from macrophages was used to establish a VSMC-macrophage indirect coculture system. Results Our results showed that both JDBM extracts and MgCl2 solutions significantly attenuated the inflammatory response stimulated by lipopolysaccharide (LPS)-activated macrophages and converted macrophages into M2-type cells. In addition, JDBM extracts and MgCl2 solutions significantly decreased the expression of genes related to VSMC phenotypic switching, migration, and proliferation in macrophages. Furthermore, the proliferation, migration, and proinflammatory phenotypic switching of VSMCs were significantly inhibited when the cells were incubated with CMs from macrophages treated with LPS + extracts or LPS + MgCl2 solutions. Conclusions Taken together, our results suggested that the magnesium in the JDBM extract could affect the functions of VSMCs through macrophage-mediated immunoregulation, inhibiting smooth muscle hyperproliferation to suppress restenosis after implantation of a biodegradable Mg-based stent.
Collapse
Affiliation(s)
- Jing Wu
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Jin
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Yun Tan
- Department of Vascular Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Xia-Fang Chen
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Neonatology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | - Guang-Yin Yuan
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.,National Engineering Research Center of Light Alloys Net Forming and State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, Shanghai, China
| | - Tong-Xin Chen
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Allergy/Immunology Innovation Team, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology/Immunology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Tan RP, Ryder I, Yang N, Lam YT, Santos M, Michael PL, Robinson DA, Ng MK, Wise SG. Macrophage Polarization as a Novel Therapeutic Target for Endovascular Intervention in Peripheral Artery Disease. JACC Basic Transl Sci 2021; 6:693-704. [PMID: 34466756 PMCID: PMC8385566 DOI: 10.1016/j.jacbts.2021.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022]
Abstract
Peripheral artery disease (PAD) has a significant impact on human health, affecting 200 million people globally. Advanced PAD severely diminishes quality of life, affecting mobility, and in its most severe form leads to limb amputation and death. Treatment of PAD is among the least effective of all endovascular procedures in terms of long-term efficacy. Chronic inflammation is a key driver of PAD; however, stents and coated balloons eluting antiproliferative drugs are most commonly used. As a result, neither stents nor coated balloons produce durable clinical outcomes in the superficial femoral artery, and both have recently been associated with significantly increased mortality. This review summarizes the most common clinical approaches and limitations to treating PAD and highlights the necessity to address the underlying causes of inflammation, identifying macrophages as a novel therapeutic target in the next generation of endovascular PAD intervention.
Collapse
Key Words
- BMS, bare-metal stent
- CAD, coronary artery disease
- DES, drug-eluting stent
- FP, femoropopliteal
- IL, interleukin
- MI, myocardial infarction
- PAD, peripheral artery disease
- PTA, percutaneous transluminal angioplasty
- SFA, superficial femoral artery
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- drug-eluting balloon
- drug-eluting stent
- endovascular intervention
- macrophage polarization
- paclitaxel
- peripheral arterial disease
- vascular healing
- vascular inflammation
Collapse
Affiliation(s)
- Richard P. Tan
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- Address for correspondence: Dr. Richard P. Tan, Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, Camperdown, Sydney, New South Wales 2006, Australia
| | - Isabelle Ryder
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Nianji Yang
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Yuen Ting Lam
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Miguel Santos
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Praveesuda L. Michael
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - David A. Robinson
- Department of Vascular Surgery, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Martin K. Ng
- Department of Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Steven G. Wise
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
Xu H, Wu Z, Jin Z, Wu X, Hu W, Liang B, Lou G, Chen Z, Yao H, Chen X, Zhou X, Xiao H, Yu C, Zhang D, Gong D, Yang L, Shi Y, Xu Y, Wang Y. Paeonol Suppresses Vasculogenesis Through Regulating Vascular Smooth Muscle Phenotypic Switching. J Endovasc Ther 2021; 29:117-131. [PMID: 34355606 DOI: 10.1177/15266028211032956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Smooth muscle cell (SMC) phenotypic switching is associated with development of a variety of occlusive vascular diseases. Paeonol has been reported to be involved in suppressing SMC proliferation. However, it is still unknown whether paeonol can regulate SMC phenotypic switching, and which eventually result in suppressing vasculogenesis. METHODS Murine left common carotid artery was injured by completely ligation, and paeonol was administrated by intraperitoneal injection. Hematoxylin and eosin (H&E) staining was performed to visualize vascular neointima formation. Rat aortic SMCs were used to determine whether paeonol suppresses cell proliferation and migration. And murine hind limb ischemia model was performed to confirm the function role of paeonol in suppressing vasculogenesis. RESULTS Complete ligation of murine common carotid artery successfully induced neointima formation. Paeonol treatment dramatically reduced the size of injury-induced neointima. Using rat aortic primary SMC, we identified that paeonol strongly suppressed cell proliferation, migration, and decreased extracellular matrix deposition. And paeonol treatment dramatically suppressed vasculogenesis after hind limb ischemia injury. CONCLUSION Paeonol could regulate SMC phenotypic switching through inhibiting proliferation and migration of SMC, which results in inhibiting ischemia-induced vasculogenesis.
Collapse
Affiliation(s)
- Huan Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziqiang Wu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhong Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Wu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wangming Hu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bing Liang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guanhua Lou
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zixian Chen
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Yao
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiongbing Chen
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zhou
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Xiao
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cenghao Yu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Delai Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Daoying Gong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lan Yang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaping Shi
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiming Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yong Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
31
|
Lee HI, Rhim WK, Kang EY, Choi B, Kim JH, Han DK. A Multilayer Functionalized Drug-Eluting Balloon for Treatment of Coronary Artery Disease. Pharmaceutics 2021; 13:614. [PMID: 33922861 PMCID: PMC8146216 DOI: 10.3390/pharmaceutics13050614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Drug-eluting balloons (DEBs) have been mostly exploited as an interventional remedy for treating atherosclerosis instead of cardiovascular stents. However, the therapeutic efficacy of DEB is limited due to their low drug delivery capability to the disease site. The aim of our study was to load drugs onto a balloon catheter with preventing drug loss during transition time and maximizing drug transfer from the surface of DEBs to the cardiovascular wall. For this, a multilayer-coated balloon catheter, composed of PVP/Drug-loaded liposome/PVP, was suggested. The hydrophilic property of 1st layer, PVP, helps to separate drug layer in hydrophilic blood vessel, and the 2nd layer with Everolimus (EVL)-loaded liposome facilitates drug encapsulation and sustained release to the targeted lesions during inflation time. Additionally, a 3rd layer with PVP can protect the inner layer during transition time for preventing drug loss. The deionized water containing 20% ethanol was utilized to hydrate EVL-loaded liposome for efficient coating processes. The coating materials showed negligible toxicity in the cells and did not induce pro-inflammatory cytokine in human coronary artery smooth muscle cells (HCASMCs), even in case of inflammation induction through LPS. The results of hemocompatibility for coating materials exhibited that protein adsorption and platelet adhesion somewhat decreased with multilayer-coated materials as compared to bare Nylon tubes. The ex vivo experiments to confirm the feasibility of further applications of multilayer-coated strategy as a DEB system demonstrated efficient drug transfer of approximately 65% in the presence of the 1st layer, to the tissue in 60 s after treatment. Taken together, a functional DEB platform with such a multilayer coating approach would be widely utilized for percutaneous coronary intervention (PCI).
Collapse
Affiliation(s)
| | | | | | | | | | - Dong-Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyenggi, Korea; (H.-I.L.); (W.-K.R.); (E.-Y.K.); (B.C.); (J.-H.K.)
| |
Collapse
|
32
|
Ling K, Zhou W, Guo Y, Hu G, Chu J, Xie F, Li Y, Wang W. H 2S attenuates oxidative stress via Nrf2/NF-κB signaling to regulate restenosis after percutaneous transluminal angioplasty. Exp Biol Med (Maywood) 2021; 246:226-239. [PMID: 32996350 PMCID: PMC7871122 DOI: 10.1177/1535370220961038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/02/2020] [Indexed: 01/05/2023] Open
Abstract
Restenosis after angioplasty of peripheral arteries is a clinical problem involving oxidative stress. Hydrogen sulfide (H2S) participates in oxidative stress regulation and activates nuclear factor erythroid 2-related factor 2 (Nrf2). This study investigated the effect of H2S and Nrf2 on restenosis-induced arterial injury. Using an in vivo rat model of restenosis, we investigated whether H2S inhibits restenosis after percutaneous transluminal angioplasty (PTA) and the oxidative stress-related mechanisms implicated therein. The involvement of Nrf2 was explored using Nrf2-shRNA. Neointimal formation and the deposition of elastic fibers were assessed histologically. Inflammatory cytokine secretion and the expression of proteins associated with oxidative stress and inflammation were evaluated. The artery of rats subjected to restenosis showed increased arterial intimal thickness, with prominent elastic fiber deposition. Sodium hydrosulfide (NaHS), an H2S donor, counteracted these changes in vivo. Restenosis caused a decrease in anti-oxidative stress signaling. This phenomenon was inhibited by NaHS, but Nrf2-shRNA counteracted the effects of NaHS. In terms of inflammation, inflammatory cytokines were upregulated, whereas NaHS suppressed the induced inflammatory reaction. Similarly, Nrf2 downregulation blocked the effect of NaHS. In vitro studies using aortic endothelial and vascular smooth muscle cells isolated from experimental animals showed consistent results as those of in vivo studies, and the participation of the nuclear factor-kappa B signaling pathway was demonstrated. Collectively, H2S played a role in regulating post-PTA restenosis by alleviating oxidative stress, modulating anti-oxidant defense, and targeting Nrf2-related pathways via nuclear factor-kappa B signaling.
Collapse
Affiliation(s)
- Ken Ling
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Zhou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi Guo
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guofu Hu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jie Chu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fen Xie
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weici Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
33
|
Wang B, Zhang M, Urabe G, Shirasu T, Guo LW, Kent KC. PERK Inhibition Promotes Post-angioplasty Re-endothelialization via Modulating SMC Phenotype Changes. J Surg Res 2021; 257:294-305. [PMID: 32871430 PMCID: PMC11034999 DOI: 10.1016/j.jss.2020.05.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/19/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Drug-eluting stents impair post-angioplasty re-endothelialization thus compromising restenosis prevention while heightening thrombotic risks. We recently found that inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) effectively mitigated both restenosis and thrombosis in rodent models. This motivated us to determine how PERK inhibition impacts re-endothelialization. METHODS Re-endothelialization was evaluated in endothelial-denuded rat carotid arteries after balloon angioplasty and periadventitial administration of PERK inhibitor in a hydrogel. To study whether PERK in smooth muscle cells (SMCs) regulates re-endothelialization by paracrinally influencing endothelial cells (ECs), denuded arteries exposing SMCs were lentiviral-infected to silence PERK; in vitro, the extracellular vesicles isolated from the medium of PDGF-activated, PERK-upregulating human primary SMCs were transferred to human primary ECs. RESULTS Treatment with PERK inhibitor versus vehicle control accelerated re-endothelialization in denuded arteries. PERK-specific silencing in the denuded arterial wall (mainly SMCs) also enhanced re-endothelialization compared to scrambled shRNA control. In vitro, while medium transfer from PDGF-activated SMCs impaired EC viability and increased the mRNA levels of dysfunctional EC markers, either PERK inhibition or silencing in donor SMCs mitigated these EC changes. Furthermore, CXCL10, a paracrine cytokine detrimental to ECs, was increased by PDGF activation and decreased after PERK inhibition or silencing in SMCs. CONCLUSIONS Attenuating PERK activity pharmacologically or genetically provides an approach to accelerating post-angioplasty re-endothelialization in rats. The mechanism may involve paracrine factors regulated by PERK in SMCs that impact neighboring ECs. This study rationalizes future development of PERK-targeted endothelium-friendly vascular interventions.
Collapse
MESH Headings
- Angioplasty, Balloon/adverse effects
- Angioplasty, Balloon/instrumentation
- Animals
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Carotid Arteries/surgery
- Coronary Restenosis/etiology
- Coronary Restenosis/prevention & control
- Disease Models, Animal
- Drug-Eluting Stents/adverse effects
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Humans
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Paracrine Communication/drug effects
- Paracrine Communication/genetics
- Protein Kinase Inhibitors/administration & dosage
- RNA, Small Interfering/metabolism
- Rats
- Re-Epithelialization/drug effects
- Re-Epithelialization/genetics
- eIF-2 Kinase/antagonists & inhibitors
- eIF-2 Kinase/genetics
Collapse
Affiliation(s)
- Bowen Wang
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Mengxue Zhang
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Cellular and Molecular Pathology Graduate Program, Department of Pathology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Go Urabe
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Takuro Shirasu
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia; Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Lian-Wang Guo
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia; Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio.
| | - K Craig Kent
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
34
|
Heinrich A, Engler MS, Güttler FV, Matthäus C, Popp J, Teichgräber UKM. Systematic evaluation of particle loss during handling in the percutaneous transluminal angioplasty for eight different drug-coated balloons. Sci Rep 2020; 10:17220. [PMID: 33057070 PMCID: PMC7560869 DOI: 10.1038/s41598-020-74227-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/29/2020] [Indexed: 11/11/2022] Open
Abstract
Paclitaxel drug coated balloons (DCBs) should provide optimal drug transfer exclusively to the target tissue. The aim of this study was to evaluate the particle loss by handling during angioplasty. A robotic arm was developed for systematic and reproducible drug abrasion experiments. The contact force on eight different commercially available DCB types was gradually increased, and high-resolution microscopic images of the deflated and inflated balloons were recorded. Three types of DCBs were classified: no abrasion of the drug in both statuses (deflated and inflated), significant abrasion only in the inflated status, and significant abrasion in both statuses. Quantitative measurements via image processing confirmed the qualitative classification and showed changes of the drug area between 2.25 and 45.73% (13.28 ± 14.29%) in the deflated status, and between 1.66 and 40.41% (21.43 ± 16.48%) in the inflated status. The structures and compositions of the DCBs are different, some are significantly more susceptible to drug loss. Particle loss by handling during angioplasty leads to different paclitaxel doses in the target regions for same DCB types. Susceptibility to involuntary drug loss may cause side effects, such as varying effective paclitaxel doses, which may explain variations in studies regarding the therapeutic outcome.
Collapse
Affiliation(s)
- Andreas Heinrich
- Department of Radiology, Jena University Hospital-Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany.
| | - Martin S Engler
- Department of Radiology, Jena University Hospital-Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Felix V Güttler
- Department of Radiology, Jena University Hospital-Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | | | - Jürgen Popp
- Leibniz Institute of Photonic Technology, 07745, Jena, Germany.,Institute of Physical Chemistry & Abbe Center of Photonics, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Ulf K-M Teichgräber
- Department of Radiology, Jena University Hospital-Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| |
Collapse
|
35
|
Giannopoulos S, Varcoe RL, Lichtenberg M, Rundback J, Brodmann M, Zeller T, Schneider PA, Armstrong EJ. Balloon Angioplasty of Infrapopliteal Arteries: A Systematic Review and Proposed Algorithm for Optimal Endovascular Therapy. J Endovasc Ther 2020; 27:547-564. [PMID: 32571125 DOI: 10.1177/1526602820931488] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Endovascular revascularization has been increasingly utilized to treat patients with chronic limb-threatening ischemia (CLTI), particularly atherosclerotic disease in the infrapopliteal arteries. Lesions of the infrapopliteal arteries are the result of 2 different etiologies: medial calcification and intimal atheromatous plaque. Although several devices are available for endovascular treatment of infrapopliteal lesions, balloon angioplasty still comprises the mainstay of therapy due to a lack of purpose-built devices. The mechanism of balloon angioplasty consists of adventitial stretching, medial necrosis, and dissection or plaque fracture. In many cases, the diffuse nature of infrapopliteal disease and plaque complexity may lead to dissection, recoil, and early restenosis. Optimal balloon angioplasty requires careful attention to assessment of vessel calcification, appropriate vessel sizing, and the use of long balloons with prolonged inflation times, as outlined in a treatment algorithm based on this systematic review. Further development of specific devices for this arterial segment are warranted, including devices for preventing recoil (eg, dedicated atherectomy devices), treating dissections (eg, tacks, stents), and preventing neointimal hyperplasia (eg, novel drug delivery techniques and drug-eluting stents). Further understanding of infrapopliteal disease, along with the development of new technologies, will help optimize the durability of endovascular interventions and ultimately improve the limb-related outcomes of patients with CLTI.
Collapse
Affiliation(s)
- Stefanos Giannopoulos
- Division of Cardiology, Rocky Mountain Regional VA Medical Center, University of Colorado, Denver, CO, USA
| | - Ramon L Varcoe
- Department of Surgery, Prince of Wales Hospital, Faculty of Medicine, University of New South Wales, The Vascular Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | | | - John Rundback
- Advanced Interventional & Vascular Services LLP, Teaneck, NJ, USA
| | - Marianne Brodmann
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Thomas Zeller
- Department of Angiology, Universitäts-Herzzentrum Bad Krozingen, Germany
| | - Peter A Schneider
- Division of Vascular and Endovascular Surgery, University of California at San Francisco, CA, USA
| | - Ehrin J Armstrong
- Division of Cardiology, Rocky Mountain Regional VA Medical Center, University of Colorado, Denver, CO, USA
| |
Collapse
|
36
|
Tepe G, Micari A, Keirse K, Zeller T, Scheinert D, Li P, Schmahl R, Jaff MR. Drug-Coated Balloon Treatment for Femoropopliteal Artery Disease: The Chronic Total Occlusion Cohort in the IN.PACT Global Study. JACC Cardiovasc Interv 2020; 12:484-493. [PMID: 30846089 DOI: 10.1016/j.jcin.2018.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/15/2018] [Accepted: 12/04/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVES This study evaluated the 12-month safety and effectiveness of a paclitaxel drug-coated balloon for treatment of intermittent claudication or rest pain in subjects with femoropopliteal chronic total occlusions (CTO). BACKGROUND CTOs are difficult to treat, and the optimal intervention remains to be determined. METHODS The IN.PACT Global Study is an international single-arm study that enrolled 1,535 patients with symptomatic femoropopliteal artery disease. The study contains prospectively defined cohorts with prospectively planned imaging analyses, including a CTO (≥5 cm) cohort in which subjects underwent duplex ultrasonography analyzed by an independent core laboratory. The primary safety endpoint was a composite of freedom from device- and procedure-related mortality through 30 days, and freedom from major target limb amputation and target vessel revascularization through 12 months. An independent Clinical Events Committee adjudicated all adverse events. The primary effectiveness endpoint was primary patency at 12 months, defined as freedom from clinically driven target lesion revascularization and freedom from restenosis. RESULTS The CTO imaging cohort had 126 subjects with 127 lesions (mean lesion length 22.83 ± 9.76 cm). Primary patency by Kaplan-Meier estimate was 85.3% through 12 months. Provisional stenting was performed in 46.8% of lesions. The primary safety composite endpoint was achieved by 88.7% of subjects. There were no device- or procedure-related deaths through 30 days or major target limb amputations through 12 months. CONCLUSIONS The paclitaxel drug-coated balloon was safe and highly effective at 12 months after treatment of subjects with CTO ≥5 cm in the femoropopliteal arteries. (IN.PACT Global Clinical Study; NCT01609296).
Collapse
Affiliation(s)
- Gunnar Tepe
- Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany.
| | - Antonio Micari
- Department of Cardiology, Humanitas Gavazzeni, Bergamo, Italy
| | - Koen Keirse
- Department of Vascular Surgery, Regional Hospital Heilig Hart Tienen, Tienen, Belgium
| | - Thomas Zeller
- Universitäts-Herzzentrum Freiburg-Bad Krozingen, Bad Krozingen, Germany
| | - Dierk Scheinert
- Division of Interventional Angiology, University Hospital Leipzig, Leipzig, Germany
| | - Pei Li
- Medtronic, Minneapolis, Minnesota
| | - Randy Schmahl
- Bakken Research Center, Medtronic, Maastricht, the Netherlands
| | | |
Collapse
|
37
|
Maleknia M, Ansari N, Haybar H, Maniati M, Saki N. Inflammatory Growth Factors and In-Stent Restenosis: Effect of Cytokines and Growth Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00240-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Kassem MM, Muqri F, Dacosta M, Bruch D, Gahtan V, Maier KG. Inhibition of heat shock protein 90 attenuates post‑angioplasty intimal hyperplasia. Mol Med Rep 2020; 21:1959-1964. [PMID: 32319637 DOI: 10.3892/mmr.2020.10994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/01/2019] [Indexed: 11/05/2022] Open
Abstract
Intimal hyperplasia (IH) is a pathologic process that leads to restenosis after treatment for peripheral arterial disease. Heat shock protein 90 (HSP90) is a molecular chaperone that regulates protein maturation. Activation of HSP90 results in increased cell migration and proliferation. 17‑N‑allylamino‑17‑demethoxygeldanamycin (17‑AAG) and 17‑dimethylaminoethylamino‑17‑demethoxygeldanamycin (17‑DMAG) are low toxicity Food and Drug Association approved HSP90 inhibitors. The current study hypothesized that HSP90 inhibition was predicted to reduce vascular smooth muscle cell (VSMC) migration and proliferation. In addition, localized HSP90 inhibition may inhibit post‑angioplasty IH formation. For proliferation, VSMCs were treated with serum‑free media (SFM), 17‑DMAG or 17‑AAG. The selected proliferative agents were SFM, platelet derived growth factor (PDGF) or fibronectin. After three days, proliferation was measured. For migration, VSMCs were treated with SFM, 17‑AAG or 17‑DMAG with SFM, PDGF or fibronectin as chemoattractants. Balloon injury to the carotid artery was performed in rats. The groups included in the present study were the control, saline control, 17‑DMAG in 20% pluronic gel delivered topically to the adventitia or intraluminal delivery of 17‑DMAG. After 14 days, arteries were fixed and sectioned for morphometric analysis. Data was analyzed using ANOVA or a student's t‑test. P<0.05 was considered to indicate a statistically significant difference. The results revealed that 17‑AAG and 17‑DMAG had no effect on cell viability. PDGF and fibronectin also increased VSMC proliferation and migration. Furthermore, both 17‑AAG and 17‑DMAG decreased cell migration and proliferation in all agonists. Topical adventitial treatment with 17‑DMAG after balloon arterial injury reduced IH. HSP90 inhibitors suppressed VSMC proliferation and migration without affecting cell viability. Topical treatment with a HSP90 inhibitor (DMAG) decreased IH formation after arterial injury. It was concluded that 17‑DMAG may be utilized as an effective therapy to prevent restenosis after revascularization.
Collapse
Affiliation(s)
- Mohammed M Kassem
- Department of Surgery, Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Furqan Muqri
- Department of Surgery, Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mary Dacosta
- College of Medicine, MD Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - David Bruch
- Department of Surgery, Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Vivian Gahtan
- Department of Surgery, Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Kristopher G Maier
- Department of Surgery, Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
39
|
Zhang B, Mo X, Yu F, Ma Y, Yan F. Ultrasound monitoring of magnet-guided delivery of mesenchymal stem cells labeled with magnetic lipid–polymer hybrid nanobubbles. Biomater Sci 2020; 8:3628-3639. [PMID: 32529995 DOI: 10.1039/d0bm00473a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells labeled with positively charged magnetic lipid–polymer hybrid nanobubbles could be tracked for magnet-guided delivery onto the site of an injured artery using ultrasound.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Ultrasound in Medicine
- Shanghai East Hospital
- Tongji University School of Medicine
- Shanghai
- China
| | - Xinhai Mo
- Department of Ultrasound in Medicine
- Shanghai East Hospital
- Tongji University School of Medicine
- Shanghai
- China
| | - Fei Yu
- Department of Ultrasound in Medicine
- Shanghai East Hospital
- Tongji University School of Medicine
- Shanghai
- China
| | - Yuqin Ma
- Department of Ultrasound
- Shenzhen Second People's Hospital
- The First Affiliated Hospital of Shenzhen University
- Shenzhen
- China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology
- Shenzhen Institute of Synthetic Biology
- Shenzhen Institutes of Advanced Technology
- Chinese Academy of Sciences
- Shenzhen
| |
Collapse
|
40
|
Zhang H, Zhang Q, Liu Y, Xue T. miR-146a and miR-146b predict increased restenosis and rapid angiographic stenotic progression risk in coronary heart disease patients who underwent percutaneous coronary intervention. Ir J Med Sci 2019; 189:467-474. [PMID: 31680203 DOI: 10.1007/s11845-019-02101-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study aimed to investigate the potential of microRNA (miR)-146a and miR-146b for predicting restenosis and rapid angiographic stenotic progression (RASP) risk in coronary heart disease (CHD) patients who underwent percutaneous coronary intervention (PCI) with drug-eluting stent (DES) implantation. METHODS In total, 255 CHD patients who underwent PCI with DES were enrolled, and their baseline, procedural, and post procedure characteristics were recorded. Plasma samples were obtained before PCI treatment to detect the miR-146a and miR-146b expression by reverse transcription quantitative polymerase chain reaction. Besides, restenosis and RASP occurrences were assessed based on coronary angiograms at 12 months after the surgery. RESULTS The occurrence rates of restenosis and RASP were 9.0% and 32.9% respectively in CHD patients who underwent PCI with DES. Furthermore, miR-146a and miR-146b expressions were elevated in CHD patients with restenosis compared with CHD patients without restenosis. Subsequent receiver operating characteristic (ROC) curve analysis showed that miR-146a (area under the curve (AUC), 0.674; 95% CI, 0.567-0.781) and miR-146b (AUC, 0.801; 95% CI, 0.729-0.875) could predict increased restenosis risk, among which miR-146b numerically exhibited a better predictive value for higher restenosis risk. Besides, miR-146a and miR-146b expressions were raised in CHD patients with RASP compared with CHD patients without RASP. Followed ROC curve analysis illuminated that miR-146a (AUC, 0.772; 95% CI, 0.714-0.829) and miR-146b (AUC, 0.706; 95% CI, 0.644-0.769) presented similar values in predicting elevated RASP risk. CONCLUSION miR-146a and miR-146b predict increased restenosis and RASP risk in CHD patients who underwent PCI with DES.
Collapse
Affiliation(s)
- Huayong Zhang
- Department of Cardiology, The Second People's Hospital of Liaocheng, Linqing, Shandong, China
| | - Qing Zhang
- Department of Cardiology, The Second People's Hospital of Liaocheng, Linqing, Shandong, China
| | - Yingchao Liu
- Department of Clinical Laboratory, The Second People's Hospital of Liaocheng, Linqing, Shandong, China
| | - Tao Xue
- Department of Cardiology, Linqing People's Hospital, 317 Yaokou Street, Xinhua Road, Linqing, 252600, Shandong, People's Republic of China.
| |
Collapse
|
41
|
Biscetti F, Ferraro PM, Hiatt WR, Angelini F, Nardella E, Cecchini AL, Santoliquido A, Pitocco D, Landolfi R, Flex A. Inflammatory Cytokines Associated With Failure of Lower-Extremity Endovascular Revascularization (LER): A Prospective Study of a Population With Diabetes. Diabetes Care 2019; 42:1939-1945. [PMID: 31371431 DOI: 10.2337/dc19-0408] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/11/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Peripheral artery disease (PAD) is one of the most relevant complications of diabetes. Although several pharmacological and revascularization approaches are available for treating patients with diabetes and PAD, an endovascular approach is often associated with postprocedural complications that can increase the risk for acute limb ischemia or amputation. However, no definitive molecular associations have been described that could explain the difference in outcomes after endovascular treatment in patients with diabetes, PAD, and chronic limb-threatening ischemia (CLTI). RESEARCH DESIGN AND METHODS We evaluated the relationship between the levels of the main cytokines associated with diabetic atherosclerosis and the outcomes after endovascular procedures in patients with diabetes, PAD, and CLTI. RESULTS A total of 299 patients with below-the-knee occlusive disease who were undergoing an angioplasty procedure were enrolled. The levels of key cytokines-osteoprotegerin (OPG), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and C-reactive protein (CRP)-were measured, and major adverse limb events (MALE) and major adverse cardiovascular events (MACE) were assessed 1, 3, 6, and 12 months after the procedure. There was a linear trend from the lowest to the highest quartile for each cytokine at baseline and incident MALE. A linear association was also observed between increasing levels of each cytokine and incident MACE. Receiver operating characteristics models were constructed using clinical and laboratory risk factors, and the inclusion of cytokines significantly improved the prediction of incident events. CONCLUSIONS We demonstrated that elevated OPG, TNF-α, IL-6, and CRP levels at baseline correlate with worse vascular outcomes in patients with diabetes, PAD, and CLTI undergoing an endovascular procedure.
Collapse
Affiliation(s)
- Federico Biscetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy .,UOC Clinica Medica e Malattie Vascolari, Rome, Italy.,Laboratorio di Biologia e Genetica Vascolare, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pietro Manuel Ferraro
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,UOC Nefrologia, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - William R Hiatt
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, and CPC Clinical Research, Aurora, CO
| | - Flavia Angelini
- Laboratorio di Biologia e Genetica Vascolare, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elisabetta Nardella
- Laboratorio di Biologia e Genetica Vascolare, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Angelo Santoliquido
- Università Cattolica del Sacro Cuore, Rome, Italy.,UOS Angiologia CIC, Rome, Italy
| | - Dario Pitocco
- Università Cattolica del Sacro Cuore, Rome, Italy.,UOSA Diabetologia, Rome, Italy
| | - Raffaele Landolfi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,UOC Clinica Medica e Malattie Vascolari, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Flex
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Laboratorio di Biologia e Genetica Vascolare, Università Cattolica del Sacro Cuore, Rome, Italy.,UOSA Medicina delle Malattie Vascolari Periferiche, Rome, Italy
| |
Collapse
|
42
|
Red Blood Cell Distribution Width Predicts 1-month Complications after Percutaneous Transluminal Angioplasty. J Med Biochem 2019; 38:468-474. [PMID: 31496911 PMCID: PMC6708289 DOI: 10.2478/jomb-2018-0047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 12/05/2018] [Indexed: 12/24/2022] Open
Abstract
Background The identification of patients at higher risk of developing percutaneous transluminal angioplasty (PTA)-related complications is pivotal for achieving better clinical outcomes. We carried out a single-center, observational, retrospective study to explore whether in-hospital changes of red blood cell distribution width (RDW) may help predicting early development of PTA-related complications. Methods The study population consisted of all consecutive patients who underwent PTA for severe peripheral artery occlusive disease (PAOD) during a 2-year period. RDW was measured at hospital admission and discharge, and the delta was calculated. Patient follow-up was routinely performed 1-month after hospital discharge, and was based on thoughtful medical assessment and arterial ultrasonography. The control population consisted of 352 ostensibly healthy subjects. Results The final PTA group consisted of 224 patients. Hemoglobin was lower, whilst mean corpuscular volume (MCV) and RDW were higher in PAOD cases than in controls. Overall, 11 PAOD patients (4.9%) developed clinically significant PTA-related complications 1-month after hospital discharge. Patients who developed 1-month PTA-related complications had lower hemoglobin concentration, but higher RDW and delta RDW than those who did not. Patients with delta RDW >1 had 60% higher risk of developing 1-month PTA-related complications and 88% higher risk of developing early reocclusion. Overall, RDW exhibited an area under the curve (AUC) of 0.68 and 0.74 for predicting 1-month PTA-related complications and early reocclusion, respectively. Conclusions The results of this study suggest that RDW may play a role for guiding the clinical decision making of PTA patients immediately after hospital discharge.
Collapse
|
43
|
Schneider PA, Laird JR, Tepe G, Brodmann M, Zeller T, Scheinert D, Metzger C, Micari A, Sachar R, Jaff MR, Wang H, Hasenbank MS, Krishnan P. Treatment Effect of Drug-Coated Balloons Is Durable to 3 Years in the Femoropopliteal Arteries: Long-Term Results of the IN.PACT SFA Randomized Trial. Circ Cardiovasc Interv 2019; 11:e005891. [PMID: 29326153 PMCID: PMC5771683 DOI: 10.1161/circinterventions.117.005891] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/29/2017] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Background— Randomized controlled trials have reported favorable 1-year outcomes with drug-coated balloons (DCBs) for the treatment of symptomatic peripheral arterial disease when compared with standard percutaneous transluminal angioplasty (PTA). Evidence remains limited on the durability of the treatment effect with DCBs in the longer term. Methods and Results— IN.PACT SFA is a single-blind, randomized trial (Randomized Trial of IN.PACT Admiral Paclitaxel-Coated Percutaneous Transluminal Angioplasty [PTA] Balloon Catheter vs Standard PTA for the Treatment of Atherosclerotic Lesions in the Superficial Femoral Artery [SFA] and/or Proximal Popliteal Artery [PPA]) that enrolled 331 patients with symptomatic (Rutherford 2–4) femoropopliteal lesions up to 18 cm in length. Patients were randomized 2:1 to receive treatment with DCB or PTA. The 36-month assessments included primary patency, freedom from clinically driven target lesion revascularization, major adverse events, and functional outcomes. At 36 months, primary patency remained significantly higher among patients treated with DCB compared with PTA (69.5% versus 45.1%; log rank P<0.001). The rates of clinically driven target lesion revascularization were 15.2% and 31.1% (P=0.002) for the DCB and PTA groups, respectively. Functional outcomes were similarly improved between treatment groups even though subjects in the DCB group required significantly fewer reinterventions versus those in the PTA group (P<0.001 for target lesion revascularization, P=0.001 for target vessel revascularization). There were no device- or procedure-related deaths as adjudicated by an independent Clinical Events Committee. Conclusions— Three-year results demonstrate a durable and superior treatment effect among patients treated with DCB versus standard PTA, with significantly higher primary patency and lower clinically driven target lesion revascularization, resulting in similar functional improvements with reduced need for repeat interventions. Clinical Trial Registration— URL: http://www.clinicaltrials.gov. Unique identifiers: NCT01175850 for IN.PACT SFA phase I in the European Union and NCT01566461 for IN.PACT SFA phase II in the United States.
Collapse
Affiliation(s)
- Peter A Schneider
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.).
| | - John R Laird
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Gunnar Tepe
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Marianne Brodmann
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Thomas Zeller
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Dierk Scheinert
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Christopher Metzger
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Antonio Micari
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Ravish Sachar
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Michael R Jaff
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Hong Wang
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Melissa S Hasenbank
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | - Prakash Krishnan
- From the Hawaii Permanente Medical Group, Kaiser Foundation Hospital, Honolulu, HI (P.A.S.); Adventist Heart and Vascular Institute, St Helena Hospital, CA (J.R.L.); Department of Diagnostic and Interventional Radiology, RoMed Klinikum, Rosenheim, Germany (G.T.); Department of Internal Medicine, Division of Angiology, Medical University, Graz, Austria (M.B.); Angiology Division, Universitäts-Herzzentrum Freiburg-Bad Krozingen, Germany (T.Z.); Department of Angiology, University Hospital Leipzig, Germany (D.S.); Wellmont Holston Valley Medical Center, Kingsport, TN (C.M.); GVM Care and Research, Maria Cecilia Hospital, Cotignola, Italy (A.M.); North Carolina Heart and Vascular, UNC-REX Health Care, Raleigh, NC (R.S.); Newton-Wellesley Hospital, MA (M.R.J.); Medtronic, Santa Rosa, CA (H.W., M.S.H.); and Icahn School of Medicine, Mount Sinai Medical Center, New York, NY (P.K.)
| | | |
Collapse
|
44
|
One step bulk modification of poly(L-lactic acid) composites with functional additives to improve mechanical and biological properties for cardiovascular implant applications. Colloids Surf B Biointerfaces 2019; 179:161-169. [PMID: 30954879 DOI: 10.1016/j.colsurfb.2019.03.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/20/2019] [Accepted: 03/29/2019] [Indexed: 01/15/2023]
Abstract
Poly(L-lactic acid) (PLLA) has been widely used as a promising biomaterial in biomedical applications due to its biodegradability and high mechanical strength. However, because of the inherent brittleness, low impact resistance, and weak thermal stability of PLLA, the modification process is usually required to utilize it for biomedical devices. Furthermore, acidic byproducts resulting from the hydrolysis of PLLA after implantation reduce the pH of the surrounding environment and cause inflammatory responses in the implanted area, leading to the failure of their clinical applications. To this end, here, we demonstrate a novel modification process for the PLLA composite with various functional additives, such as cis-aconitic anhydride (AA), triacetin (TA), isosorbide derivative (ISB), and/or Pluronic® F127 (F). The modified PLLA composite with TA and F (PLLA/TF) showed significantly improved elongation at break and Young's modulus and retained tensile strength. Moreover, incorporating magnesium hydroxide (MH) nanoparticles (PLLA/TFMH) significantly reduced acid-induced inflammation responses caused by the acidic degradation products of PLLA. Reduced plasma protein adsorption was observed in the PLLA/TFMH. These results suggest that the one step bulk modification of biodegradable PLLA using TA, F, and MH will have great potential in cardiovascular implant applications.
Collapse
|
45
|
The Yin and Yang of carbon nanomaterials in atherosclerosis. Biotechnol Adv 2018; 36:2232-2247. [PMID: 30342084 DOI: 10.1016/j.biotechadv.2018.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/06/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
With unique characteristics such as high surface area, capacity of various functionalization, low weight, high conductivity, thermal and chemical stability, and free radical scavenging, carbon nanomaterials (CNMs) such as carbon nanotubes (CNTs), fullerene, graphene (oxide), carbon nanohorns (CNHs), and their derivatives have increasingly been utilized in nanomedicine and biomedicine. On the one hand, owing to ever-increasing applications of CNMs in technological and industrial fields as well as presence of combustion-derived CNMs in the ambient air, the skepticism has risen over the adverse effects of CNMs on human being. The influences of CNMs on cardiovascular system and cardiovascular diseases (CVDs) such as atherosclerosis, of which consequences are ischemic heart disease and ischemic stroke, as the main causes of death, is of paramount importance. In this regard, several studies have been devoted to specify the biomedical applications and cardiovascular toxicity of CNMs. Therefore, the aim of this review is to specify the roles and applications of various CNMs in atherosclerosis, and also identify the key role playing parameters in cardiovascular toxicity of CNMs so as to be a clue for prospective deployment of CNMs.
Collapse
|
46
|
Fukasawa M, Isobe M, Nanto S, Nakamura M, Haruguchi H, Miyake T, Morishita R. NF-κB Decoy Oligodeoxynucleotide-Coated Balloon Catheter for Arteriovenous Fistula in Hemodialysis. Kidney Int Rep 2018; 4:126-138. [PMID: 30596176 PMCID: PMC6308821 DOI: 10.1016/j.ekir.2018.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022] Open
Abstract
Introduction New treatments to inhibit neointimal formation after percutaneous transluminal angioplasty (PTA) are needed for patients undergoing chronic hemodialysis (HD). We compared the efficacy and safety of AMG0102, a balloon catheter containing nuclear factor κB (NF-κB) decoy oligodeoxynucleotide (ODN) with the PTA balloon catheter (control group) for arteriovenous fistula (AVF) stenosis. Methods In total, 175 patients (age ≥20 years, undergoing HD, with venous stenosis at the anastomotic region) were registered in this prospective open-label, randomized study. Patients were followed postoperatively for 36 weeks. The duration of primary patency on the targeted venous stenosis site (primary endpoint) was estimated by the Kaplan–Meier method. Results A lower restenosis risk was observed for the AMG0102 group, but it was not statistically significant (stratified log-rank test P = 0.250, hazard ratio [HR] 0.774; 95% confidence interval [CI]: 0.500–1.198). The median duration of primary patency was 245 days and 172 days in the AMG0102 and control groups, respectively. After stratification based on the status of diabetes complications, the HR was 0.666 (95% CI: 0.366–1.212; P = 0.183) and the median duration of primary patency was prolonged by 108 days in the AMG0102 group with diabetes complications (245 days) compared with the control group (137 days). Adverse event (AE) incidence up to 36 postoperative weeks did not differ between groups. Four device failures occurred in 3 patients (AMG0102 group), but none resulted in AEs. Conclusion Further modifications to enhance NF-κB decoy ODN uptake and efficacy are necessary to show its clinical utility for AVF stenosis in chronic HD.
Collapse
Affiliation(s)
- Mizuya Fukasawa
- Division of Integrated Renal Replacement Therapy, Department of Urology, University of Yamanashi, Yamanashi, Japan
- Correspondence: Mizuya Fukasawa, Division of Integrated Renal Replacement Therapy, Department of Urology, University of Yamanashi, Yamanashi, Japan.
| | | | - Shinsuke Nanto
- Department of Cardiology, Nishinomiya Municipal Central Hospital, Hyogo, Japan
| | - Masato Nakamura
- Division of Cardiovascular Medicine, Toho University, Ohashi Medical Center, Tokyo, Japan
| | | | - Takashi Miyake
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
47
|
Abstract
Endovascular intervention is a mainstay treatment of peripheral artery disease (PAD) in addition to aggressive risk factor modification and exercise programs in patients with favorable anatomy or in those who are considered too high risk for surgical intervention. Treatment with percutaneous transluminal angioplasty (PTA) and bare metal stents (BMS) has been limited by high rates of in-stent restenosis (ISR) requiring repeat revascularization. Drug-eluting stents (DES), developed and designed to reduce ISR, offer a promising solution to the current challenges in endovascular management of PAD. Several randomized clinical trials have shown improved short- and mid-term outcomes with DES as compared with both PTA and BMS. Herein we provide an up-to-date review of the current literature on DES use in PAD.
Collapse
|
48
|
Wang D, Uhrin P, Mocan A, Waltenberger B, Breuss JM, Tewari D, Mihaly-Bison J, Huminiecki Ł, Starzyński RR, Tzvetkov NT, Horbańczuk J, Atanasov AG. Vascular smooth muscle cell proliferation as a therapeutic target. Part 1: molecular targets and pathways. Biotechnol Adv 2018; 36:1586-1607. [PMID: 29684502 DOI: 10.1016/j.biotechadv.2018.04.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases are a major cause of human death worldwide. Excessive proliferation of vascular smooth muscle cells contributes to the etiology of such diseases, including atherosclerosis, restenosis, and pulmonary hypertension. The control of vascular cell proliferation is complex and encompasses interactions of many regulatory molecules and signaling pathways. Herein, we recapitulated the importance of signaling cascades relevant for the regulation of vascular cell proliferation. Detailed understanding of the mechanism underlying this process is essential for the identification of new lead compounds (e.g., natural products) for vascular therapies.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria; Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Hațieganu" University of Medicine and Pharmacy, Strada Gheorghe Marinescu 23, 400337 Cluj-Napoca, Romania; Institute for Life Sciences, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal, 263136 Nainital, Uttarakhand, India
| | - Judit Mihaly-Bison
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Łukasz Huminiecki
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Rafał R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, 1618 Sofia, Bulgaria
| | - Jarosław Horbańczuk
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| |
Collapse
|
49
|
Mollenhauer M, Mehrkens D, Rudolph V. Nitrated fatty acids in cardiovascular diseases. Nitric Oxide 2018; 78:S1089-8603(17)30292-6. [PMID: 29588164 DOI: 10.1016/j.niox.2018.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death and accounts for one third of disease-related mortality worldwide. Dysregulated redox mechanisms, in particular the formation of reactive oxygen species (ROS) play a pivotal pathogenetic role in CVD. Nitro-fatty acids (NO2-FAs) are electrophilic molecules which have a NO2-group bound to one of their olefinic carbons. They are endogenously formed by the reaction of reactive nitrogen species with unsaturated fatty acids. Basal levels of NO2-FAs are in the low nanomolar range and higher concentrations can be encountered under acidic (stomach) and inflammatory (e.g. ischemia/reperfusion) conditions. Dietary intake of polyunsaturated fatty acids in combination with nitrites raises circulating NO2-FAs to a clinically relevant level in mice. NO2-FAs undergo reversible covalent binding to cysteine residues and by virtue of these posttranslational protein modifications act as potent anti-inflammatory signaling mediators via modulation of various critical pathways like nuclear factor E2-related factor 2 (Nrf2)- and peroxisome proliferator-activated receptor γ (PPARγ) activation, nuclear factor-kappa B (NF-κB) inhibition and hem oxygenase-1 (HO-1)- and heat shock protein (HSP) induction. In this review article, we summarize recent findings about the effects and underlying molecular mechanisms of NO2-FAs from a variety of pre-clinical cardiovascular disease models. The described findings suggest the potential of NO2-FAs to emerge as therapeutic agents with a broad range of potential clinical applications for CVD.
Collapse
Affiliation(s)
- Martin Mollenhauer
- Department of Cardiology, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Dennis Mehrkens
- Department of Cardiology, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Volker Rudolph
- Department of Cardiology, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
50
|
Li H, Gui H, Yuan G, Zheng X, Gao C, Yuan H. Increased plasma olfactomedin 2 after interventional therapy is a predictor for restenosis in lower extremity arteriosclerosis obliterans patients. Scandinavian Journal of Clinical and Laboratory Investigation 2018; 78:269-274. [PMID: 29553861 DOI: 10.1080/00365513.2018.1452287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Animal studies have indicated that olfactomedin 2 (OLFM2) is involved in the process of vascular remolding. The aim of the present study was to investigate circulating OLFM2 levels in lower extremity arteriosclerosis obliterans (LEASO) patients and the association of OLFM2 with postoperative restenosis in patients. A total of 203 LEASO patients were enrolled in the present study. Plasma OLFM2 was measured before and 6 h after interventional therapy. After 6 months, patients were divided into a restenosis group and a non-restenosis group. Inter-group and intra-group differences in plasma OLFM2 were compared. The correlation between plasma OLFM2 and the severity of restenosis was analyzed by Spearman's correlation analysis. An receiver operating characteristic (ROC) curve was used to evaluate the predictive efficacy of plasma OLFM2 on restenosis. Logistic regression was used to determine the risk factors for restenosis. Postoperative OLFM2 in the restenosis group was significantly higher compared with the non-restenosis group (34.07 ± 5.76 ng/mL vs. 19.53 ± 2.99 ng/mL). No significant difference in preoperative plasma OLFM2 levels was identified between the two groups (10.92 ± 2.49 ng/mL vs. 11.54 ± 3.18 ng/mL). Postoperative OLFM2 levels were positively correlated with the severity of restenosis (r = 0.728, p < .001). The area under the ROC curve was 0.902 (95% confidence interval (CI): 0.874-0.965), with a cutoff value of 26.91 ng/mL (95% CI: 26.16-28.32). Plasma OLFM2 was an independent risk factor for restenosis. Our results suggest that plasma OLFM2 is a potential biomarker for restenosis and may be a novel target for the treatment of restenosis.
Collapse
Affiliation(s)
- Hongbo Li
- a Department of Vascular Surgery , People's Hospital of Shouguang , Weifang , Shandong , China.,b Department of Vascular Surgery , Shandong Provincial Hospital , Jinan , Shandong , China
| | - Hua Gui
- c Department of Surgery , People's Hospital of Shouguang , Weifang , Shandong , China
| | - Guohong Yuan
- c Department of Surgery , People's Hospital of Shouguang , Weifang , Shandong , China
| | - Xiaomei Zheng
- a Department of Vascular Surgery , People's Hospital of Shouguang , Weifang , Shandong , China
| | - Changkuan Gao
- d Department of Clinical Laboratory , Qilu Hospital , Jinan , Shandong , China
| | - Hai Yuan
- b Department of Vascular Surgery , Shandong Provincial Hospital , Jinan , Shandong , China
| |
Collapse
|