1
|
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
|
2
|
Alanazi YA, Al‐kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Negm WA, AlAnazi FH, Alrouji M, Batiha GE. Role of Autophagy in Type 2 Diabetes Mellitus: The Metabolic Clash. J Cell Mol Med 2024; 28:e70240. [PMID: 39656379 PMCID: PMC11629865 DOI: 10.1111/jcmm.70240] [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: 12/12/2023] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is developed due to the development of insulin resistance (IR) and pancreatic β cell dysfunction with subsequent hyperglycaemia. Hyperglycaemia-induced oxidative stress and endoplasmic reticulum (ER) stress enhances inflammatory disorders, leading to further pancreatic β cell dysfunction. These changes trigger autophagy activation, which recycles cytoplasmic components and injured organelles. Autophagy regulates pancreatic β cell functions by different mechanisms. Though the exact role of autophagy in T2DM is not completely elucidated, that could be beneficial or detrimental. Therefore, this review aims to discuss the exact role of autophagy in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Yousef Abud Alanazi
- Department of Pediatrics, College of MedicineMajmaah UniversityMajmaahSaudi Arabia
| | - Haydar M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentFunogenAthensGreece
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Mostafa M. Bahaa
- Pharmacy Practice Department, Faculty of PharmacyHorus UniversityNew DamiettaEgypt
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of PharmacyTanta UniversityTantaEgypt
| | - Faisal Holil AlAnazi
- Department of Internal Medicine, College of MedicineMajmaah UniversityMajmaahSaudi Arabia
| | - Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesShaqra UniversityShaqraSaudi Arabia
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
3
|
Ning L, Zanella S, Tomov ML, Amoli MS, Jin L, Hwang B, Saadeh M, Chen H, Neelakantan S, Dasi LP, Avazmohammadi R, Mahmoudi M, Bauser‐Heaton HD, Serpooshan V. Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400476. [PMID: 38696618 PMCID: PMC11234432 DOI: 10.1002/advs.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/09/2024] [Indexed: 05/04/2024]
Abstract
Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.
Collapse
Affiliation(s)
- Liqun Ning
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of Mechanical EngineeringCleveland State UniversityClevelandOH44115USA
| | - Stefano Zanella
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Martin L. Tomov
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Linqi Jin
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Boeun Hwang
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Maher Saadeh
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Huang Chen
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Sunder Neelakantan
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lakshmi Prasad Dasi
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Reza Avazmohammadi
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
- J. Mike Walker ’66 Department of Mechanical EngineeringTexas A&M UniversityCollege StationTX77840USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LandingMI48824USA
| | - Holly D. Bauser‐Heaton
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
- Sibley Heart Center at Children's Healthcare of AtlantaAtlantaGA30322USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
| |
Collapse
|
4
|
Rutecki S, Pakuła-Iwańska M, Leśniewska-Bocianowska A, Matuszewska J, Rychlewski D, Uruski P, Stryczyński Ł, Naumowicz E, Szubert S, Tykarski A, Mikuła-Pietrasik J, Książek K. Mechanisms of carboplatin- and paclitaxel-dependent induction of premature senescence and pro-cancerogenic conversion of normal peritoneal mesothelium and fibroblasts. J Pathol 2024; 262:198-211. [PMID: 37941520 DOI: 10.1002/path.6223] [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: 05/22/2023] [Revised: 09/08/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023]
Abstract
Carboplatin (CPT) and paclitaxel (PCT) are the optimal non-surgical treatment of epithelial ovarian cancer (EOC). Although their growth-restricting influence on EOC cells is well known, their impact on normal peritoneal cells, including mesothelium (PMCs) and fibroblasts (PFBs), is poorly understood. Here, we investigated whether, and if so, by what mechanism, CPT and PCT induce senescence of omental PMCs and PFBs. In addition, we tested whether PMC and PFB exposure to the drugs promotes the development of a pro-cancerogenic phenotype. The results showed that CPT and PCT induce G2/M growth arrest-associated senescence of normal peritoneal cells and that the strongest induction occurs when the drugs act together. PMCs senesce telomere-independently with an elevated p16 level and via activation of AKT and STAT3. In PFBs, telomeres shorten along with an induction of p21 and p53, and their senescence proceeds via the activation of ERK1/2. Oxidative stress in CPT + PCT-treated PMCs and PFBs is extensive and contributes causatively to their premature senescence. Both PMCs and PFBs exposed to CPT + PCT fuel the proliferation, migration, and invasion of established (A2780, OVCAR-3, SKOV-3) and primary EOCs, and this activity is linked with an overproduction of multiple cytokines altering the cancer cell transcriptome and controlled by p38 MAPK, NF-κB, STAT3, Notch1, and JAK1. Collectively, our findings indicate that CPT and PCT lead to iatrogenic senescence of normal peritoneal cells, which paradoxically and opposing therapeutic needs alters their phenotype towards pro-cancerogenic. It cannot be excluded that these adverse outcomes of chemotherapy may contribute to EOC relapse in the case of incomplete tumor eradication and residual disease initiation. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
- Poznań University of Medical Sciences Doctoral School, Poznań, Poland
| | | | | | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Łukasz Stryczyński
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Eryk Naumowicz
- General Surgery Ward, Medical Centre HCP, Poznań, Poland
| | - Sebastian Szubert
- Department of Gynecology, Division of Gynecologic Oncology, Poznań University of Medical Sciences, Poznań, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
5
|
Lee S, Yoon CH, Oh DH, Anh TQ, Jeon KH, Chae IH, Park KD. Gelatin microgel-coated balloon catheter with enhanced delivery of everolimus for long-term vascular patency. Acta Biomater 2024; 173:314-324. [PMID: 37949201 DOI: 10.1016/j.actbio.2023.11.001] [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: 05/17/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
In-stent restenosis (ISR) after percutaneous coronary intervention is a major reason for limited long-term patency due to complex neointimal proliferation caused by vascular injury. Drug-coated balloon (DCB) has been developed to treat various cardiovascular diseases including ISR by providing anti-proliferative drugs into blood vessel tissues. However, a significant proportion of the drug is lost during balloon tracking, resulting in ineffective drug delivery to the target region. In this study, we report an everolimus-coated balloon (ECB) using everolimus-loaded gelatin-hydroxyphenyl propionic acid microgel (GM) with enhanced everolimus delivery to vascular walls for long-term patency. GM with high drug loading (> 97%) was simply prepared by homogenizing enzyme-mediated crosslinked hydrogels. The optimal condition to prepare GM-coated ECB (GM-ECB) was established by changing homogenization time and ethanol solvent concentration (30 ∼ 80%). In vitro sustained everolimus release for 30 d, and cellular efficacy using smooth muscle cells and vascular endothelial cells were evaluated. Additionally, an in vivo drug transfer levels of GM-ECB using rabbit femoral arteries were assessed with reduced drug loss and efficient drug delivery capability. Finally, using ISR-induced porcine models, effective in vivo vascular patency 4 weeks after treatment of ECBs was also confirmed. Thus, this study strongly demonstrates that GM can be used as a potential drug delivery platform for DCB application. STATEMENT OF SIGNIFICANCE: We report an ECB using everolimus-loaded GM prepared by homogenization of enzymatic cross-linked hydrogel. GM showed efficient drug loading (> 97 %) and controllable size. GM-ECB exhibited potential to deliver everolimus in a sustained manner to target area with drug efficacy and viability against SMC and EC. Although GM-ECB had much lower drug content compared to controls, animal study demonstrated enhanced drug transfer and reduced drug loss of GM-ECB due to the protection of encapsulated drugs by GM, and the possible interaction between GM and endothelium. Finally, vascular patency and safety were assessed using ISR-induced porcine models. We suggest an advanced DCB strategy to alleviate rapid drug clearance by bloodstream while improving drug delivery for a long-term vascular patency.
Collapse
Affiliation(s)
- Simin Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Chang-Hwan Yoon
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dong Hwan Oh
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Tu Quang Anh
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Ki-Hyun Jeon
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In-Ho Chae
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| | - Ki Dong Park
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
6
|
Mia MAR, Dey D, Sakib MR, Biswas MY, Prottay AAS, Paul N, Rimti FH, Abdullah Y, Biswas P, Iftehimul M, Paul P, Sarkar C, El-Nashar HAS, El-Shazly M, Islam MT. The efficacy of natural bioactive compounds against prostate cancer: Molecular targets and synergistic activities. Phytother Res 2023; 37:5724-5754. [PMID: 37786304 DOI: 10.1002/ptr.8017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/17/2023] [Accepted: 09/05/2023] [Indexed: 10/04/2023]
Abstract
Globally, prostate cancer (PCa) is regarded as a challenging health issue, and the number of PCa patients continues to rise despite the availability of effective treatments in recent decades. The current therapy with chemotherapeutic drugs has been largely ineffective due to multidrug resistance and the conventional treatment has restricted drug accessibility to malignant tissues, necessitating a higher dosage resulting in increased cytotoxicity. Plant-derived bioactive compounds have recently attracted a great deal of attention in the field of PCa treatment due to their potent effects on several molecular targets and synergistic effects with anti-PCa drugs. This review emphasizes the molecular mechanism of phytochemicals on PCa cells, the synergistic effects of compound-drug interactions, and stem cell targeting for PCa treatment. Some potential compounds, such as curcumin, phenethyl-isothiocyanate, fisetin, baicalein, berberine, lutein, and many others, exert an anti-PCa effect via inhibiting proliferation, metastasis, cell cycle progression, and normal apoptosis pathways. In addition, multiple studies have demonstrated that the isolated natural compounds: d-limonene, paeonol, lanreotide, artesunate, and bicalutamide have potential synergistic effects. Further, a significant number of natural compounds effectively target PCa stem cells. However, further high-quality studies are needed to firmly establish the clinical efficacy of these phytochemicals against PCa.
Collapse
Affiliation(s)
- Md Abdur Rashid Mia
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Dipta Dey
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Musfiqur Rahman Sakib
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Md Yeaman Biswas
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology (JUST), Jashore, Bangladesh
| | - Abdullah Al Shamsh Prottay
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Niloy Paul
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Fahmida Hoque Rimti
- Bachelor of Medicine and Surgery, Chittagong Medical College, Chawkbazar, Bangladesh
| | - Yusuf Abdullah
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Partha Biswas
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology (JUST), Jashore, Bangladesh
| | - Md Iftehimul
- Department of Fisheries and Marine Bioscience, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Priyanka Paul
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Chandan Sarkar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, Bangladesh
| |
Collapse
|
7
|
Zieschang AD, Hoffseth KF, Dugas TR, Astete CE, Boldor D. Method for assessing coating uniformity of angioplasty balloons coated with poly(lactic-co-glycolic acid) nanoparticles loaded with quercetin. PARTICULATE SCIENCE AND TECHNOLOGY 2023; 42:601-611. [PMID: 38966520 PMCID: PMC11221576 DOI: 10.1080/02726351.2023.2269881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
In this study, we aim to quantify coating uniformity and correlate fluorescence intensity to drug loading for drug-coated angioplasty balloons (DCB) coated with 5, 10, 15, or 20 layers of poly(lactic-co-glycolic acid) nanoparticles (NPs) entrapped with quercetin. Uniformity was quantified from histograms and horizontal line profiles of microscopic fluorescent images acquired with sample specific parameters, and cracks in the coating were measured and counted. The fluorescence of images acquired with global parameters was correlated with quercetin loading measured via gravimetric/HPLC analysis. More layers on DCBs may be associated with less uniform coatings, as indicated by differences in histogram standard deviations. The line profile percent deviation from average for each sample was <20%. Cracks were present on all balloons, but their length was not significantly different between samples. The 5-layer DCBs had the fewest cracks, whereas the 15-layer DCBs had the most cracks. A strong positive correlation (R = 0.896) was identified between fluorescence intensity and drug loading. A relationship between the number of layers and coating uniformity seems to exist, but further investigations are required for confirmation. Fluorescence intensity appears to strongly predict drug loading, demonstrating that fluorescent imaging may be a viable alternative to drug release studies.
Collapse
Affiliation(s)
- Allison D. Zieschang
- Louisiana State University and the LSU AgCenter, Department of Biological and Agricultural Engineering, Baton Rouge, Louisiana, United States of America
| | - Kevin F. Hoffseth
- Louisiana State University and the LSU AgCenter, Department of Biological and Agricultural Engineering, Baton Rouge, Louisiana, United States of America
| | - Tammy R. Dugas
- Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Baton Rouge, Louisiana, United States of America
| | - Carlos E. Astete
- Louisiana State University and the LSU AgCenter, Department of Biological and Agricultural Engineering, Baton Rouge, Louisiana, United States of America
| | - Dorin Boldor
- Louisiana State University and the LSU AgCenter, Department of Biological and Agricultural Engineering, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
8
|
Elhadad S, Redmond D, Huang J, Tan A, Laurence J. MASP2 inhibition by narsoplimab suppresses endotheliopathies characteristic of transplant-associated thrombotic microangiopathy: in vitro and ex vivo evidence. Clin Exp Immunol 2023; 213:252-264. [PMID: 37191586 PMCID: PMC10361744 DOI: 10.1093/cei/uxad055] [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: 03/07/2023] [Revised: 04/17/2023] [Accepted: 05/15/2023] [Indexed: 05/17/2023] Open
Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) is an endotheliopathy complicating up to 30% of allogeneic hematopoietic stem cell transplants (alloHSCT). Positive feedback loops among complement, pro-inflammatory, pro-apoptotic, and coagulation cascade likely assume dominant roles at different disease stages. We hypothesized that mannose-binding lectin-associated serine protease 2 (MASP2), principal activator of the lectin complement system, is involved in the microvascular endothelial cell (MVEC) injury characteristic of TA-TMA through pathways that are susceptible to suppression by anti-MASP2 monoclonal antibody narsoplimab. Pre-treatment plasmas from 8 of 9 TA-TMA patients achieving a complete TMA response in a narsoplimab clinical trial activated caspase 8, the initial step in apoptotic injury, in human MVEC. This was reduced to control levels following narsoplimab treatment in 7 of the 8 subjects. Plasmas from 8 individuals in an observational TA-TMA study, but not 8 alloHSCT subjects without TMA, similarly activated caspase 8, which was blocked in vitro by narsoplimab. mRNA sequencing of MVEC exposed to TA-TMA or control plasmas with and without narsoplimab suggested potential mechanisms of action. The top 40 narsoplimab-affected transcripts included upregulation of SerpinB2, which blocks apoptosis by inactivating procaspase 3; CHAC1, which inhibits apoptosis in association with mitigation of oxidative stress responses; and pro-angiogenesis proteins TM4SF18, ASPM, and ESM1. Narsoplimab also suppressed transcripts encoding pro-apoptotic and pro-inflammatory proteins ZNF521, IL1R1, Fibulin-5, aggrecan, SLC14A1, and LOX1, and TMEM204, which disrupts vascular integrity. Our data suggest benefits to narsoplimab use in high-risk TA-TMA and provide a potential mechanistic basis for the clinical efficacy of narsoplimab in this disorder.
Collapse
Affiliation(s)
- Sonia Elhadad
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, New York, NY, USA
| | - Jenny Huang
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, New York, NY, USA
| | - Adrian Tan
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Jeffrey Laurence
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
9
|
Santovito D, Steffens S, Barachini S, Madonna R. Autophagy, innate immunity, and cardiac disease. Front Cell Dev Biol 2023; 11:1149409. [PMID: 37234771 PMCID: PMC10206260 DOI: 10.3389/fcell.2023.1149409] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Autophagy is an evolutionarily conserved mechanism of cell adaptation to metabolic and environmental stress. It mediates the disposal of protein aggregates and dysfunctional organelles, although non-conventional features have recently emerged to broadly extend the pathophysiological relevance of autophagy. In baseline conditions, basal autophagy critically regulates cardiac homeostasis to preserve structural and functional integrity and protect against cell damage and genomic instability occurring with aging. Moreover, autophagy is stimulated by multiple cardiac injuries and contributes to mechanisms of response and remodeling following ischemia, pressure overload, and metabolic stress. Besides cardiac cells, autophagy orchestrates the maturation of neutrophils and other immune cells, influencing their function. In this review, we will discuss the evidence supporting the role of autophagy in cardiac homeostasis, aging, and cardioimmunological response to cardiac injury. Finally, we highlight possible translational perspectives of modulating autophagy for therapeutic purposes to improve the care of patients with acute and chronic cardiac disease.
Collapse
Affiliation(s)
- Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Unit of Milan, Institute for Genetic and Biomedical Research (IRGB), National Research Council, Milan, Italy
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Serena Barachini
- Hematology Division, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rosalinda Madonna
- Cardiology Division, Cardio-Thoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Surgical, Medical, Molecular Pathology & Critical Care Sciences, University of Pisa, Pisa, Italy
| |
Collapse
|
10
|
Zieschang AD, Hoffseth KF, Dugas TR, Astete CE, Boldor D. Method for assessing coating uniformity of angioplasty balloons coated with poly(lactic-co-glycolic acid) nanoparticles loaded with quercetin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524614. [PMID: 36711445 PMCID: PMC9882345 DOI: 10.1101/2023.01.18.524614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Significance Drug-coated angioplasty balloons (DCBs) are used to treat peripheral artery disease, and proper dosage depends on coating characteristics like uniformity and number of layers. Aim Quantify coating uniformity and correlate fluorescence intensity to drug loading for DCBs coated with 5, 10, 15, or 20 layers of poly(lactic-co-glycolic acid) nanoparticles (NPs) entrapped with quercetin. Approach Images of DCBs were acquired using fluorescence microscopy. Coating uniformity was quantified from histograms and horizontal line profiles, and cracks on the balloons were measured and counted. Fluorescence intensity was correlated with the drug loading of quercetin found from gravimetric analysis coupled with high-performance liquid chromatography (HPLC). Results Higher numbers of coating layers on DCBs may be associated with less uniform coatings. Cracks in the coating were present on all balloons, and the length of cracks was not significantly different between balloons coated with different numbers of layers or balloons coated with the same number of layers. A strong positive correlation was identified between fluorescence intensity and drug loading. Conclusion There may be a relationship between the number of NP layers and the uniformity of the coating, but further investigation is needed to confirm this. Fluorescence intensity appears to be a strong predictor of drug loading on DCBs coated with quercetin-entrapped NPs, demonstrating that fluorescent imaging may be a viable alternative to drug release studies.
Collapse
|
11
|
Xie X, Wu Q, Liu Y, Chen C, Chen Z, Xie C, Song M, Jiang Z, Qi X, Liu S, Tang Z, Wu Z. Vascular endothelial growth factor attenuates neointimal hyperplasia of decellularized small-diameter vascular grafts by modulating the local inflammatory response. Front Bioeng Biotechnol 2022; 10:1066266. [PMID: 36605251 PMCID: PMC9808043 DOI: 10.3389/fbioe.2022.1066266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Small-diameter vascular grafts (diameter <6 mm) are in high demand in clinical practice. Neointimal hyperplasia, a common complication after implantation of small-diameter vascular grafts, is one of the common causes of graft failure. Modulation of local inflammatory responses is a promising strategy to attenuates neointimal hyperplasia. Vascular endothelial growth factor (VEGF) is an angiogenesis stimulator that also induces macrophage polarization and modulates inflammatory responses. In the present study, we evaluated the effect of VEGF on the neointima hyperplasia and local inflammatory responses of decellularized vascular grafts. In the presence of rhVEGF-165 in RAW264.6 macrophage culture, rhVEGF-165 induces RAW264.6 macrophage polarization to M2 phenotype. Decellularized bovine internal mammary arteries were implanted into the subcutaneous and infrarenal abdominal aorta of New Zealand rabbits, with rhVEGF-165 applied locally to the adventitial of the grafts. The vascular grafts were removed en-bloc and submitted to histological and immunofluorescence analyses on days 7 and 28 following implantation. The thickness of the fibrous capsule and neointima was thinner in the VEGF group than that in the control group. In the immunofluorescence analysis, the number of M2 macrophages and the ratio of M2/M1 macrophages in vascular grafts in the VEGF group were higher than those in the control group, and the proinflammatory factor IL-1 was expressed less than in the control group, but the anti-inflammatory factor IL-10 was expressed more. In conclusion, local VEGF administration attenuates neointimal hyperplasia in decellularized small-diameter vascular grafts by inducing macrophage M2 polarization and modulating the inflammatory response.
Collapse
Affiliation(s)
- Xinlong Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Cardiothoracic Surgery, The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qiying Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuhong Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunyang Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeguo Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingzhe Song
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenlin Jiang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoke Qi
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sixi Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, Hunan, China
| | - Zhongshi Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, Hunan, China,*Correspondence: Zhongshi Wu,
| |
Collapse
|
12
|
Ursino H, Zhang B, Ludtka C, Webb A, Allen JB. Hemocompatibility of all-trans retinoic acid-loaded citrate polymer coatings for vascular stents. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022; 8:579-592. [PMID: 36714809 PMCID: PMC9881644 DOI: 10.1007/s40883-022-00257-y] [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: 07/28/2021] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 02/02/2023]
Abstract
Purpose Current strategies implementing drug-eluting polymer stent coatings fail to fully address the lasting effects of endothelial suppression which ultimately result in delayed reendothelialization and thrombogenic complications. The present study investigates the in vitro hemocompatibility of all-trans retinoic acid loaded poly (1,8-octanediol-co-citrate) coatings (AtRA-POC coatings) for advanced intravascular stent technology. The ability of these materials in supporting endothelial restoration via migration and proliferation while inhibiting smooth muscle cell growth is also explored. Methods Using in vitro models, the hemocompatibility of AtRA-loaded POC-coated cobalt chromium (CoCr) vascular stents was evaluated in terms of platelet and inflammatory activity. Platelet activity was quantified by platelet adhesion and platelet activation, further supported by SEM visualization. Inflammatory activity was quantified by the production of proinflammatory cytokines by THP1 monocytes. Lastly, in vitro wound healing and an 5-Ethynyl-2'deoxyuridine (EdU) and pico green DNA assays were used in quantitating endothelial and smooth muscle cell migration and proliferation. Results Experimental examinations of platelet adhesion and activation demonstrate significant reductions in the platelet response to POC coated AtRA loaded stents when compared to bare CoCr stents. Such findings reveal AtRA-POC coatings to have significantly improved hemocompatibility compared to that of bare metal stents and at least as good as POC alone. Similarly, in reference to LPS-stimulated controls, Human monocyte-like THP1 cells in culture with AtRA-POC-CoCr stents for 24 hours showed reduced detection of proinflammatory cytokines, comparable to that of bare CoCr and untreated controls. This result supports AtRA-POC coatings as possessing limited immunological potential. Observations from in vitro endothelial and smooth muscle cell investigations demonstrate the ability of the drug AtRA to allow cell processes involved in restoration of the endothelium while inhibiting smooth muscle cell processes. Conclusion This study demonstrates AtRA loaded POC coatings are hemocompatible, noninflammatory, and provide a promising strategy in enhancing vascular stent techniques and clinical integration. Possessing hemocompatibility and immunological compatibility that is at least as good as bare metal stents as clinical standards support the use of AtRA-POC coatings for vascular applications. Additionally, selectively reducing smooth muscle cell proliferation while supporting endothelial cell proliferation and migration further demonstrates the potential of these materials in significantly improving the state of vascular stent technology in the area of stent thrombosis and neointimal hyperplasia.
Collapse
Affiliation(s)
- Heather Ursino
- Univeristy of Florida, Materials Science and Engineering, Gainesville, FL, USA
| | - Bisheng Zhang
- Univeristy of Florida, Materials Science and Engineering, Gainesville, FL, USA
| | | | - Antonio Webb
- Univeristy of Florida, Materials Science and Engineering, Gainesville, FL, USA
| | - Josephine B. Allen
- Univeristy of Florida, Materials Science and Engineering, Gainesville, FL, USA
- Univeristy of Florida, Biomedical Engineering, Gainesville, FL, USA
| |
Collapse
|
13
|
Zhao F, Satyanarayana G, Zhang Z, Zhao J, Ma XL, Wang Y. Endothelial Autophagy in Coronary Microvascular Dysfunction and Cardiovascular Disease. Cells 2022; 11:2081. [PMID: 35805165 PMCID: PMC9265562 DOI: 10.3390/cells11132081] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023] Open
Abstract
Coronary microvascular dysfunction (CMD) refers to a subset of structural and/or functional disorders of coronary microcirculation that lead to impaired coronary blood flow and eventually myocardial ischemia. Amid the growing knowledge of the pathophysiological mechanisms and the development of advanced tools for assessment, CMD has emerged as a prevalent cause of a broad spectrum of cardiovascular diseases (CVDs), including obstructive and nonobstructive coronary artery disease, diabetic cardiomyopathy, and heart failure with preserved ejection fraction. Of note, the endothelium exerts vital functions in regulating coronary microvascular and cardiac function. Importantly, insufficient or uncontrolled activation of endothelial autophagy facilitates the pathogenesis of CMD in diverse CVDs. Here, we review the progress in understanding the pathophysiological mechanisms of autophagy in coronary endothelial cells and discuss their potential role in CMD and CVDs.
Collapse
Affiliation(s)
- Fujie Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | | | - Zheng Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (F.Z.); (Z.Z.); (J.Z.); (X.-L.M.)
| |
Collapse
|
14
|
Montero P, Milara J, Pérez-Leal M, Estornut C, Roger I, Pérez-Fidalgo A, Sanz C, Cortijo J. Paclitaxel-Induced Epidermal Alterations: An In Vitro Preclinical Assessment in Primary Keratinocytes and in a 3D Epidermis Model. Int J Mol Sci 2022; 23:ijms23031142. [PMID: 35163066 PMCID: PMC8834980 DOI: 10.3390/ijms23031142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Paclitaxel is a microtubule-stabilizing chemotherapeutic agent approved for the treatment of ovarian, non-small cell lung, head, neck, and breast cancers. Despite its beneficial effects on cancer and widespread use, paclitaxel also damages healthy tissues, including the skin. However, the mechanisms that drive these skin adverse events are not clearly understood. In the present study, we demonstrated, by using both primary epidermal keratinocytes (NHEK) and a 3D epidermis model, that paclitaxel impairs different cellular processes: paclitaxel increased the release of IL-1α, IL-6, and IL-8 inflammatory cytokines, produced reactive oxygen species (ROS) release and apoptosis, and reduced the endothelial tube formation in the dermal microvascular endothelial cells (HDMEC). Some of the mechanisms driving these adverse skin events in vitro are mediated by the activation of toll-like receptor 4 (TLR-4), which phosphorylate transcription of nuclear factor kappa B (NF-κb). This is the first study analyzing paclitaxel effects on healthy human epidermal cells with an epidermis 3D model, and will help in understanding paclitaxel's effects on the skin.
Collapse
Affiliation(s)
- Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.E.); (I.R.); (C.S.); (J.C.)
- Correspondence: (P.M.); (J.M.); Tel.: +34-963864631 (P.M.)
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.E.); (I.R.); (C.S.); (J.C.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
- Pharmacy Unit, University General Hospital Consortium, 46014 Valencia, Spain
- Correspondence: (P.M.); (J.M.); Tel.: +34-963864631 (P.M.)
| | - Martín Pérez-Leal
- Faculty of Health Sciences, Universidad Europea de Valencia, 46010 Valencia, Spain;
| | - Cristina Estornut
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.E.); (I.R.); (C.S.); (J.C.)
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.E.); (I.R.); (C.S.); (J.C.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
| | - Alejandro Pérez-Fidalgo
- Department of Medical Oncology, University Clinic Hospital of Valencia, 46010 Valencia, Spain;
- Biomedical Research Networking Centre on Cancer (CIBERONC), Health Institute Carlos III, 28029 Madrid, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Celia Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.E.); (I.R.); (C.S.); (J.C.)
- Health Sciences, Pre-Departmental Section of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.E.); (I.R.); (C.S.); (J.C.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
- Research and Teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain
| |
Collapse
|
15
|
Tierney JW, Evans BC, Cheung-Flynn J, Wang B, Colazo JM, Polcz ME, Cook RS, Brophy CM, Duvall CL. Therapeutic MK2 inhibition blocks pathological vascular smooth muscle cell phenotype switch. JCI Insight 2021; 6:e142339. [PMID: 34622803 PMCID: PMC8525639 DOI: 10.1172/jci.insight.142339] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Vascular procedures, such as stenting, angioplasty, and bypass grafting, often fail due to intimal hyperplasia (IH), wherein contractile vascular smooth muscle cells (VSMCs) dedifferentiate to synthetic VSMCs, which are highly proliferative, migratory, and fibrotic. Previous studies suggest MAPK-activated protein kinase 2 (MK2) inhibition may limit VSMC proliferation and IH, although the molecular mechanism underlying the observation remains unclear. We demonstrated here that MK2 inhibition blocked the molecular program of contractile to synthetic dedifferentiation and mitigated IH development. Molecular markers of the VSMC contractile phenotype were sustained over time in culture in rat primary VSMCs treated with potent, long-lasting MK2 inhibitory peptide nanopolyplexes (MK2i-NPs), a result supported in human saphenous vein specimens cultured ex vivo. RNA-Seq of MK2i-NP-treated primary human VSMCs revealed programmatic switching toward a contractile VSMC gene expression profile, increasing expression of antiinflammatory and contractile-associated genes while lowering expression of proinflammatory, promigratory, and synthetic phenotype-associated genes. Finally, these results were confirmed using an in vivo rabbit vein graft model where brief, intraoperative treatment with MK2i-NPs decreased IH and synthetic phenotype markers while preserving contractile proteins. These results support further development of MK2i-NPs as a therapy for blocking VSMC phenotype switch and IH associated with cardiovascular procedures.
Collapse
Affiliation(s)
- J. William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian C. Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bo Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Monica E. Polcz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of General Surgery and
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Colleen M. Brophy
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
16
|
Eckert MA, Orozco C, Xiao J, Javellana M, Lengyel E. The Effects of Chemotherapeutics on the Ovarian Cancer Microenvironment. Cancers (Basel) 2021; 13:3136. [PMID: 34201616 PMCID: PMC8268261 DOI: 10.3390/cancers13133136] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/31/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is characterized by a complex and dynamic tumor microenvironment (TME) composed of cancer-associated fibroblasts (CAFs), immune cells, endothelial cells, and adipocytes. Although most approved therapies target cancer cells, a growing body of evidence suggests that chemotherapeutic agents have an important role in regulating the biology of the diverse cells that compose the TME. Understanding how non-transformed cells respond and adapt to established therapeutics is necessary to completely comprehend their action and develop novel therapeutics that interrupt undesired tumor-stroma interactions. Here, we review the effects of chemotherapeutic agents on normal cellular components of the host-derived TME focusing on CAFs. We concentrate on therapies used in the treatment of HGSOC and synthesize findings from studies focusing on other cancer types and benign tissues. Agents such as platinum derivatives, taxanes, and PARP inhibitors broadly affect the TME and promote or inhibit the pro-tumorigenic roles of CAFs by modifying the bidirectional cross-talk between tumor and stromal cells in the tumor organ. While most chemotherapy research focuses on cancer cells, these studies emphasize the need to consider all cell types within the tumor organ when evaluating chemotherapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (M.A.E.); (C.O.); (J.X.); (M.J.)
| |
Collapse
|
17
|
Heparin-Tagged PLA-PEG Copolymer-Encapsulated Biochanin A-Loaded (Mg/Al) LDH Nanoparticles Recommended for Non-Thrombogenic and Anti-Proliferative Stent Coating. Int J Mol Sci 2021; 22:ijms22115433. [PMID: 34063962 PMCID: PMC8196732 DOI: 10.3390/ijms22115433] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Drug-eluting stents have been widely implanted to prevent neointimal hyperplasia associated with bare metal stents. Conventional polymers and anti-proliferative drugs suffer from stent thrombosis due to the non-selective nature of the drugs and hypersensitivity to polymer degradation products. Alternatively, various herbal anti-proliferative agents are sought, of which biochanin A (an isoflavone phytoestrogen) was known to have anti-proliferative and vasculoprotective action. PLA-PEG diblock copolymer was tagged with heparin, whose degradation releases heparin locally and prevents thrombosis. To get a controlled drug release, biochanin A was loaded in layered double hydroxide nanoparticles (LDH), which are further encapsulated in a heparin-tagged PLA-PEG copolymer. LDH nanoparticles are synthesized by a co-precipitation process; in situ as well as ex situ loading of biochanin A were done. PLA-PEG-heparin copolymer was synthesized by esterification reaction, and the drug-loaded nanoparticles are coated. The formulation was characterized by FTIR, XRD, DSC, DLS, and TEM. In vitro drug release studies, protein adhesion, wettability, hemocompatibility, and degradation studies were performed. The drug release was modeled by mathematical models to further emphasize the mechanism of drug release. The developed drug-eluting stent coating is non-thrombogenic, and it offers close to zero-order release for 40 days, with complete polymer degradation in 14 weeks.
Collapse
|
18
|
Cannabidiol Promotes Endothelial Cell Survival by Heme Oxygenase-1-Mediated Autophagy. Cells 2020; 9:cells9071703. [PMID: 32708634 PMCID: PMC7407143 DOI: 10.3390/cells9071703] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
Cannabidiol (CBD), a non-psychoactive cannabinoid, has been reported to mediate antioxidant, anti-inflammatory, and anti-angiogenic effects in endothelial cells. This study investigated the influence of CBD on the expression of heme oxygenase-1 (HO-1) and its functional role in regulating metabolic, autophagic, and apoptotic processes of human umbilical vein endothelial cells (HUVEC). Concentrations up to 10 µM CBD showed a concentration-dependent increase of HO-1 mRNA and protein and an increase of the HO-1-regulating transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). CBD-induced HO-1 expression was not decreased by antagonists of cannabinoid-activated receptors (CB1, CB2, transient receptor potential vanilloid 1), but by the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). The incubation of HUVEC with 6 µM CBD resulted in increased metabolic activity, while 10 µM CBD caused decreased metabolic activity and an induction of apoptosis, as demonstrated by enhanced caspase-3 cleavage. In addition, CBD triggered a concentration-dependent increase of the autophagy marker LC3A/B-II. Both CBD-induced LC3A/B-II levels and caspase-3 cleavage were reduced by NAC. The inhibition of autophagy by bafilomycin A1 led to apoptosis induction by 6 µM CBD and a further increase of the proapoptotic effect of 10 µM CBD. On the other hand, the inhibition of HO-1 activity with tin protoporphyrin IX (SnPPIX) or knockdown of HO-1 expression by Nrf2 siRNA was associated with a decrease in CBD-mediated autophagy and apoptosis. In summary, our data show for the first time ROS-mediated HO-1 expression in endothelial cells as a mechanism by which CBD mediates protective autophagy, which at higher CBD concentrations, however, can no longer prevent cell death inducing apoptosis.
Collapse
|
19
|
Forouzanfar F, Mousavi SH. Targeting Autophagic Pathways by Plant Natural Compounds in Cancer Treatment. Curr Drug Targets 2020; 21:1237-1249. [PMID: 32364070 DOI: 10.2174/1389450121666200504072635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/26/2020] [Accepted: 03/19/2020] [Indexed: 12/29/2022]
Abstract
Nowadays, natural compounds of plant origin with anticancer effects have gained more attention because of their clinical safety and broad efficacy profiles. Autophagy is a multistep lysosomal degradation pathway that may have a unique potential for clinical benefit in the setting of cancer treatment. To retrieve articles related to the study, the databases of Google Scholar, Web of sciences, Medline and Scopus, using the following keywords: Autophagic pathways; herbal medicine, oncogenic autophagic pathways, tumor-suppressive autophagic pathways, and cancer were searched. Although natural plant compounds such as resveratrol, curcumin, oridonin, gossypol, and paclitaxel have proven anticancer potential via autophagic signaling pathways, there is still a great need to find new natural compounds and investigate the underlying mechanisms, to facilitate their clinical use as potential anticancer agents through autophagic induction.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Yu D, Gernapudi R, Drucker C, Sarkar R, Ucuzian A, Monahan TS. The myristoylated alanine-rich C kinase substrate differentially regulates kinase interacting with stathmin in vascular smooth muscle and endothelial cells and potentiates intimal hyperplasia formation. J Vasc Surg 2019; 70:2021-2031.e1. [PMID: 30929966 PMCID: PMC6765458 DOI: 10.1016/j.jvs.2018.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/06/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Restenosis limits the durability of all cardiovascular reconstructions. Vascular smooth muscle cell (VSMC) proliferation drives this process, but an intact, functional endothelium is necessary for vessel patency. Current strategies to prevent restenosis employ antiproliferative agents that affect both VSMCs and endothelial cells (ECs). Knockdown of the myristoylated alanine-rich C kinase substrate (MARCKS) arrests VSMC proliferation and paradoxically potentiates EC proliferation. MARCKS knockdown decreases expression of the kinase interacting with stathmin (KIS), increasing p27kip1 expression, arresting VSMC proliferation. Here, we seek to determine how MARCKS influences KIS protein expression in these two cell types. METHODS Primary human coronary artery VSMCs and ECs were used for in vitro experiments. MARCKS was depleted by transfection with small interfering RNA. Messenger RNA was quantitated with the real-time reverse transcription polymerase chain reaction. Protein expression was determined by Western blot analysis. Ubiquitination was determined with immunoprecipitation. MARCKS and KIS binding was assessed with co-immunoprecipitation. Intimal hyperplasia was induced in CL57/B6 mice with a femoral artery wire injury. MARCKS was knocked down in vivo by application of 10 μM of small interfering RNA targeting MARCKS suspended in 30% Pluronic F-127 gel. Intimal hyperplasia formation was assessed by measurement of the intimal thickness on cross sections of the injured artery. Re-endothelialization was determined by quantitating the binding of Evans blue dye to the injured artery. RESULTS MARCKS knockdown did not affect KIS messenger RNA expression in either cell type. In the presence of cycloheximide, MARCKS knockdown in VSMCs decreased KIS protein stability but had no effect in ECs. The effect of MARCKS knockdown on KIS stability was abrogated by the 26s proteasome inhibitor MG-132. MARCKS binds to KIS in VSMCs but not in ECs. MARCKS knockdown significantly increased the level of ubiquitinated KIS in VSMCs but not in ECs. MARCKS knockdown in vivo resulted in decreased KIS expression. Furthermore, MARCKS knockdown in vivo resulted in decreased 5-ethynyl-2'-deoxyuridine integration and significantly reduced intimal thickening. MARCKS knockdown enhanced endothelial barrier function recovery 4 days after injury. CONCLUSIONS MARCKS differentially regulates the KIS protein stability in VSMCs and ECs. The difference in stability is due to differential ubiquitination of KIS in these two cell types. The differential interaction of MARCKS and KIS provides a possible explanation for the observed difference in ubiquitination. The effect of MARCKS knockdown on KIS expression persists in vivo, potentiates recovery of the endothelium, and abrogates intimal hyperplasia formation.
Collapse
Affiliation(s)
- Dan Yu
- Department of Surgery, Baltimore Veterans Affairs Medical Center, Baltimore, Md; Department of Surgery, University of Maryland School of Medicine, Baltimore, Md
| | | | - Charles Drucker
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Md
| | - Rajabrata Sarkar
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Md; Department of Physiology, University of Maryland School of Medicine, Baltimore, Md
| | - Areck Ucuzian
- Department of Surgery, Baltimore Veterans Affairs Medical Center, Baltimore, Md; Department of Surgery, University of Maryland School of Medicine, Baltimore, Md
| | - Thomas S Monahan
- Department of Surgery, Baltimore Veterans Affairs Medical Center, Baltimore, Md; Department of Surgery, University of Maryland School of Medicine, Baltimore, Md.
| |
Collapse
|
21
|
Flentje A, Kalsi R, Monahan TS. Small GTPases and Their Role in Vascular Disease. Int J Mol Sci 2019; 20:ijms20040917. [PMID: 30791562 PMCID: PMC6413073 DOI: 10.3390/ijms20040917] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Over eighty million people in the United States have cardiovascular disease that can affect the heart causing myocardial infarction; the carotid arteries causing stroke; and the lower extremities leading to amputation. The treatment for end-stage cardiovascular disease is surgical—either endovascular therapy with balloons and stents—or open reconstruction to reestablish blood flow. All interventions damage or destroy the protective inner lining of the blood vessel—the endothelium. An intact endothelium is essential to provide a protective; antithrombotic lining of a blood vessel. Currently; there are no agents used in the clinical setting that promote reendothelialization. This process requires migration of endothelial cells to the denuded vessel; proliferation of endothelial cells on the denuded vessel surface; and the reconstitution of the tight adherence junctions responsible for the formation of an impermeable surface. These processes are all regulated in part and are dependent on small GTPases. As important as the small GTPases are for reendothelialization, dysregulation of these molecules can result in various vascular pathologies including aneurysm formation, atherosclerosis, diabetes, angiogenesis, and hypertension. A better understanding of the role of small GTPases in endothelial cell migration is essential to the development for novel agents to treat vascular disease.
Collapse
Affiliation(s)
- Alison Flentje
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Richa Kalsi
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Thomas S Monahan
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| |
Collapse
|
22
|
Mikuła-Pietrasik J, Witucka A, Pakuła M, Uruski P, Begier-Krasińska B, Niklas A, Tykarski A, Książek K. Comprehensive review on how platinum- and taxane-based chemotherapy of ovarian cancer affects biology of normal cells. Cell Mol Life Sci 2019; 76:681-697. [PMID: 30382284 PMCID: PMC6514066 DOI: 10.1007/s00018-018-2954-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/24/2022]
Abstract
One of the most neglected aspects of chemotherapy are changes, and possible consequences of these changes, that occur in normal somatic cells. In this review, we summarize effects of selected drugs used to treat ovarian cancer (platin derivatives-cisplatin and carboplatin; and taxanes-paclitaxel and docetaxel) on cellular metabolism, acquisition of reactive stroma features, cellular senescence, inflammatory reactions, apoptosis, autophagy, mitophagy, oxidative stress, DNA damage, and angiogenesis in various types of normal cells, including fibroblasts, epithelial cells, endothelial cells, and neurons. The activity of these drugs against the normal cells is presented from a broader perspective of their desirable anti-tumoral effects.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Anna Witucka
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Martyna Pakuła
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Beata Begier-Krasińska
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznań, Poland.
| |
Collapse
|
23
|
Jin Z, Chen S, Wu H, Wang J, Wang L, Gao W. Inhibition of autophagy after perforator flap surgery increases flap survival and angiogenesis. J Surg Res 2018; 231:83-93. [PMID: 30278973 DOI: 10.1016/j.jss.2018.05.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 04/02/2018] [Accepted: 05/16/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND The survival ratio of multiterritory perforator flap is variable. Therefore, surviving mechanisms are increasingly explored to identify novel therapeutics. The condition of the choke zone is essential for perforator flap survival. In this study, we investigated autophagy in the choke zone after flap surgery. MATERIALS AND METHODS The flap model involved a perforator flap with three territories that was located on the right dorsal side of a rat. A total of 36 rats were divided into six groups, including the control, 0 d postoperative (PO), 1, 3, 5, and 7 d PO groups. In addition, 72 rats were divided into three groups, including a control group, a 3-methyladenine (3-MA) group, and a rapamycin group. Skin tissue of rats was used for measuring autophagy proteins, vascular endothelial growth factor (VEGF) expression, and histological examination. On day 7 after surgery, the survival ratio of each flap was determined. RESULTS The expression of autophagy and VEGF in the second choke zone (choke II) was increased after flap surgery. Among the three groups, the survival ratio of flaps in the 3-MA group was the highest. Furthermore, the angiogenesis level in the 3-MA group in choke II was the highest among the three groups. CONCLUSIONS Autophagy was initiated by surgery in choke II, and VEGF expression in choke II was increased after flap surgery. Inhibiting autophagy after perforator flap surgery is beneficial for flap survival and for promoting angiogenesis in choke II.
Collapse
Affiliation(s)
- Zhicheng Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shao Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hongqiang Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jieke Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Long Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
24
|
Jiang F. Autophagy in vascular endothelial cells. Clin Exp Pharmacol Physiol 2017; 43:1021-1028. [PMID: 27558982 DOI: 10.1111/1440-1681.12649] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/30/2016] [Accepted: 08/08/2016] [Indexed: 12/18/2022]
Abstract
The importance of autophagy in cardiovascular physiology and cardiovascular disease is increasingly recognized; however, the precise biological effects and underlying mechanisms of autophagy in the cardiovascular system are still poorly understood. In the last few years, the effects of autophagy in endothelial cells have attracted great interests. This article provides a summary of our current knowledge on the regulatory factors, signalling mechanisms, and functional outcomes of autophagy in endothelial cells. It is suggested that in most situations, induction of an autophagic response has cytoprotective effects. The beneficial effects of autophagy in endothelial cells are likely to be context-dependent, since autophagy may also contribute to cell death under certain circumstances. In addition to regulating endothelial cell survival or death, autophagy is also involved in modulating other important functions, such as nitric oxide production, angiogenesis and haemostasis/thrombosis. The mounting data will help us draw a clear picture of the roles of autophagy in endothelial cell biology and dysfunction. Given the pivotal role of endothelial dysfunction in the pathogenesis of vascular disease, disruptions of autophagy in endothelial cells are likely to have significant contributions. This is supported by some preliminary ex vivo data indicating that compromised autophagic functions may be important in the development of endothelial dysfunctions associated with diabetes and ageing.
Collapse
Affiliation(s)
- Fan Jiang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
25
|
Sachdev U, Lotze MT. Perpetual change: autophagy, the endothelium, and response to vascular injury. J Leukoc Biol 2017; 102:221-235. [PMID: 28626046 PMCID: PMC6608075 DOI: 10.1189/jlb.3ru1116-484rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Current studies of vascular health, aging, and autophagy emphasize how the endothelium adapts to stress and contributes to disease. The endothelium is far from an inert barrier to blood-borne cells, pathogens, and chemical signals; rather, it actively translates circulating mediators into tissue responses, changing rapidly in response to physiologic stressors. Macroautophagy-the cellular ingestion of effete organelles and protein aggregates to provide anabolic substrates to fuel bioenergetics in times of stress-plays an important role in endothelial cell homeostasis, vascular remodeling, and disease. These roles include regulating vascular tone, sustaining or limiting cell survival, and contributing to the development of atherosclerosis secondary to infection, inflammation, and angiogenesis. Autophagy modulates these critical functions of the endothelium in a dynamic and perpetual response to tissue and intravascular cues.
Collapse
Affiliation(s)
- Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
Zhang YQ, Tian F, Chen JS, Chen YD, Zhou Y, Li B, Ma Q, Zhang Y. Delayed reendothelialization with rapamycin is rescued by the addition of nicorandil in balloon-injured rat carotid arteries. Oncotarget 2016; 7:75926-75939. [PMID: 27713157 PMCID: PMC5342788 DOI: 10.18632/oncotarget.12444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 09/24/2016] [Indexed: 01/09/2023] Open
Abstract
Rapamycin is an immunosuppressive agent that is added to drug eluting stents. It prevents restenosis, but it also impairs reendothelialization. Nicorandil is a hybrid agent with adenosine triphosphated (ATP)-sensitive K+ (KATP) channel opener and nitrate properties. It prevents oxidative stress and cell apoptosis induced by rapamycin in endothelial cells in vitro. However, whether nicorandil promotes reendothelialization after angioplasty delayed by rapamycin remains to be determined. Balloon injury model was established in SD rats. Nicorandil increased reendothelialization impaired by rapamycin, and it decreased xanthine oxidase (XO)-generated reactive oxygen species (ROS) induced by rapamycin. In addition, eNOS expression inhibited by rapamycin was increased by nicorandil in vivo. In vitro, rapamycin-impeded cardiac microvascular endothelial cells (CMECs) migration, proliferation and rapamycin-induced ROS production were reversed by nicorandil. Knockdown of XO partially inhibited rapamycin-induced ROS production and cell apoptosis in CMECs, and it promoted CMECs migration and proliferation suppressed by rapamycin. Knockdown of Akt partially prevents eNOS upregulation promoted by nicorandil. The beneficial effect of nicorandil is exhibited by inhibiting XO and up-regulating Akt pathway. Nicorandil combined with rapamycin in effect rescue the deficiencies of rapamycin alone in arterial healing after angioplasty.
Collapse
Affiliation(s)
- Ying Qian Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jin Song Chen
- Department of Cardiology, Chinese PLA 175th Hospital, Fujian, China.,Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yun Dai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Ying Zhou
- VIP Medical Service Department, Beijing Shijitan Hospital, Beijing, China.,Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Bo Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
27
|
Brahmbhatt A, Misra S. Techniques in Vascular and Interventional Radiology Drug Delivery Technologies in the Superficial Femoral Artery. Tech Vasc Interv Radiol 2016; 19:145-52. [PMID: 27423996 DOI: 10.1053/j.tvir.2016.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Peripheral arterial disease (PAD) affects over 8 million people in the United States alone. Although great strides have been made in reducing the burden of cardiovascular disease the prevalence of PAD is expected to rise with the age of global population. PAD characterized by narrowing of arterial blood can be asymptomatic or cause limb threatening claudication. It has been classically treated with bypass, but these techniques have been supplanted by endovascular therapy. Plain old balloon angioplasty has been successful in helping revascularize lesions, but its effect has not been durable because of restenosis. This prompted the creation of several technologies aimed at reducing restenosis. These advances slowly improved outcomes and the durability of endovascular management. Among the main tools used in current endovascular practice are drug-delivery devices aimed at inhibiting the inflammatory and proliferative pathways that lead to restenosis. This article examines the current drug-delivery technologies used in the superficial femoral artery.
Collapse
Affiliation(s)
- Akshaar Brahmbhatt
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology Mayo Clinic, Rochester, MN; Department of Radiology, Mayo Clinic, Rochester, MN; Rutgers-New Jersey Medical School, Newark, NJ
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology Mayo Clinic, Rochester, MN; Department of Radiology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
28
|
Gu J, Hu W, Song ZP, Chen YG, Zhang DD, Wang CQ. Rapamycin Inhibits Cardiac Hypertrophy by Promoting Autophagy via the MEK/ERK/Beclin-1 Pathway. Front Physiol 2016; 7:104. [PMID: 27047390 PMCID: PMC4796007 DOI: 10.3389/fphys.2016.00104] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/03/2016] [Indexed: 01/07/2023] Open
Abstract
Rapamycin, also known as sirolimus, is an antifungal agent and immunosuppressant drug used to prevent organ rejection in transplantation. However, little is known about the role of rapamycin in cardiac hypertrophy and the signaling pathways involved. Here, the effect of rapamycin was examined using phenylephrine (PE) induced cardiomyocyte hypertrophy in vitro and in a rat model of aortic banding (AB) - induced hypertrophy in vivo. Inhibition of MEK/ERK signaling reversed the effect of rapamycin on the up-regulation of LC3-II, Beclin-1 and Noxa, and the down-regulation of Mcl-1 and p62. Silencing of Noxa or Beclin-1 suppressed rapamycin-induced autophagy, and co-immunoprecipitation experiments showed that Noxa abolishes the inhibitory effect of Mcl-1 on Beclin-1, promoting autophagy. In vivo experiments showed that rapamycin decreased AB-induced cardiac hypertrophy in a MEK/ERK dependent manner. Taken together, our results indicate that rapamycin attenuates cardiac hypertrophy by promoting autophagy through a mechanism involving the modulation of Noxa and Beclin-1 expression by the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Jun Gu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China; Department of Cardiology, Shanghai Minhang Hospital, Fudan UniversityShanghai, China
| | - Wei Hu
- Department of Cardiology, Shanghai Minhang Hospital, Fudan University Shanghai, China
| | - Zhi-Ping Song
- Department of Cardiology, Shanghai Minhang Hospital, Fudan University Shanghai, China
| | - Yue-Guang Chen
- Department of Cardiology, Shanghai Minhang Hospital, Fudan University Shanghai, China
| | - Da-Dong Zhang
- Department of Cardiology, Shanghai Minhang Hospital, Fudan University Shanghai, China
| | - Chang-Qian Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine Shanghai, China
| |
Collapse
|
29
|
Endothelial Repair and Regeneration Following Intimal Injury. J Cardiovasc Transl Res 2016; 9:91-101. [DOI: 10.1007/s12265-016-9677-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/13/2016] [Indexed: 12/19/2022]
|
30
|
Killinger BA, Moszczynska A. Epothilone D prevents binge methamphetamine-mediated loss of striatal dopaminergic markers. J Neurochem 2015; 136:510-25. [PMID: 26465779 DOI: 10.1111/jnc.13391] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 09/12/2015] [Accepted: 09/21/2015] [Indexed: 02/06/2023]
Abstract
Exposure to binge methamphetamine (METH) can result in a permanent or transient loss of dopaminergic (DAergic) markers such as dopamine (DA), dopamine transporter, and tyrosine hydroxylase (TH) in the striatum. We hypothesized that the METH-induced loss of striatal DAergic markers was, in part, due to a destabilization of microtubules (MTs) in the nigrostriatal DA pathway that ultimately impedes anterograde axonal transport of these markers. To test this hypothesis, adult male Sprague-Dawley rats were treated with binge METH or saline in the presence or absence of epothilone D (EpoD), a MT-stabilizing compound, and assessed 3 days after the treatments for the levels of several DAergic markers as well as for the levels of tubulins and their post-translational modifications (PMTs). Binge METH induced a loss of stable long-lived MTs within the striatum but not within the substantia nigra pars compacta (SNpc). Treatment with a low dose of EpoD increased the levels of markers of stable MTs and prevented METH-mediated deficits in several DAergic markers in the striatum. In contrast, administration of a high dose of EpoD appeared to destabilize MTs and potentiated the METH-induced deficits in several DAergic markers. The low-dose EpoD also prevented the METH-induced increase in striatal DA turnover and increased behavioral stereotypy during METH treatment. Together, these results demonstrate that MT dynamics plays a role in the development of METH-induced losses of several DAergic markers in the striatum and may mediate METH-induced degeneration of terminals in the nigrostriatal DA pathway. Our study also demonstrates that MT-stabilizing drugs such as EpoD have a potential to serve as useful therapeutic agents to restore function of DAergic nerve terminals following METH exposure when administered at low doses. Administration of binge methamphetamine (METH) negatively impacts neurotransmission in the nigrostriatal dopamine (DA) system. The effects of METH include decreasing the levels of DAergic markers in the striatum. We have determined that high-dose METH destabilizes microtubules in this pathway, which is manifested by decreased levels of acetylated (Acetyl) and detyrosinated (Detyr) α-tubulin (I). A microtubule stabilizing agent epothilone D protects striatal microtubules form the METH-induced loss of DAergic markers (II). These findings provide a new strategy for protection form METH - restoration of proper axonal transport.
Collapse
Affiliation(s)
- Bryan A Killinger
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
31
|
MARCKS Signaling Differentially Regulates Vascular Smooth Muscle and Endothelial Cell Proliferation through a KIS-, p27kip1- Dependent Mechanism. PLoS One 2015; 10:e0141397. [PMID: 26528715 PMCID: PMC4631550 DOI: 10.1371/journal.pone.0141397] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
Background Overexpression of the myristolated alanine-rich C kinase substrate (MARCKS) occurs in vascular proliferative diseases such as restenosis after bypass surgery. MARCKS knockdown results in arrest of vascular smooth muscle cell (VSMC) proliferation with little effect on endothelial cell (EC) proliferation. We sought to identify the mechanism of differential regulation by MARCKS of VSMC and EC proliferation in vitro and in vivo. Methods and Results siRNA-mediated MARCKS knockdown in VSMCs inhibited proliferation and prevented progression from phase G0/G1 to S. Protein expression of the cyclin-dependent kinase inhibitor p27kip1, but not p21cip1 was increased by MARCKS knockdown. MARCKS knockdown did not affect proliferation in VSMCs derived from p27kip1-/- mice indicating that the effect of MARCKS is p27kip1-dependent. MARCKS knockdown resulted in decreased phosphorylation of p27kip1 at threonine 187 and serine 10 as well as, kinase interacting with stathmin (KIS), cyclin D1, and Skp2 expression. Phosphorylation of p27kip1 at serine 10 by KIS is required for nuclear export and degradation of p27kip1. MARCKS knockdown caused nuclear trapping of p27kip1. Both p27kip1 nuclear trapping and cell cycle arrest were released by overexpression of KIS, but not catalytically inactive KIS. In ECs, MARCKS knockdown paradoxically increased KIS expression and cell proliferation. MARCKS knockdown in a murine aortic injury model resulted in decreased VSMC proliferation determined by bromodeoxyuridine (BrdU) integration assay, and inhibition of vascular wall thickening. MARCKS knockdown increased the rate of re-endothelialization. Conclusions MARCKS knockdown arrested VSMC cell cycle by decreasing KIS expression. Decreased KIS expression resulted in nuclear trapping of p27kip1 in VSMCs. MARCKS knockdown paradoxically increased KIS expression in ECs resulting in increased EC proliferation. MARCKS knockdown significantly attenuated the VSMC proliferative response to vascular injury, but accelerated reestablishment of an intact endothelium. MARCKS is a novel translational target with beneficial cell type-specific effects on both ECs and VSMCs.
Collapse
|
32
|
Hu N, Kong LS, Chen H, Li WD, Qian AM, Wang XY, Du XL, Li CL, Yu XB, Li XQ. Autophagy protein 5 enhances the function of rat EPCs and promotes EPCs homing and thrombus recanalization via activating AKT. Thromb Res 2015; 136:642-51. [DOI: 10.1016/j.thromres.2015.06.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/18/2015] [Accepted: 06/30/2015] [Indexed: 12/13/2022]
|
33
|
Spira D, Grözinger G, Domschke N, Bantleon R, Schmehl J, Wiskirchen J, Wiesinger B. Cell Cycle Regulation of Smooth Muscle Cells--Searching for Inhibitors of Neointima Formation: Is Combretastatin A4 an Alternative to Sirolimus and Paclitaxel? J Vasc Interv Radiol 2015; 26:1388-95. [PMID: 26169455 DOI: 10.1016/j.jvir.2015.05.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/04/2015] [Accepted: 05/10/2015] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To compare the effects of sirolimus, paclitaxel, and combretastatin A4 (CA4) on regulatory proteins of the cell cycle in proliferating smooth muscle cells (SMCs). MATERIALS AND METHODS Human aortic SMCs were treated with sirolimus, paclitaxel, and CA4 at 5 × 10(-9) mol/L. After 1 day, half of the cells were harvested (DAY1 group). The treatment medium of the other half was replaced with culture medium on day 4, and those cells were harvested on day 5 (DAY5 group). Cyclins D1, D2, E, and A and cyclin-dependent kinase (CDK) inhibitors p16, p21, and p27 were detected by Western blot technique. Quantification was performed by scanning densitometry of the specific bands. RESULTS In the DAY1 group, treatment with sirolimus resulted in decreased intracellular levels of cyclins D2 and A (P < .05). Increased D cyclins and reduced levels of cyclins E and A (P < .05) in the DAY5 group indicated a permanent G1/S block by sirolimus. Paclitaxel led to only slight alterations of cyclin and CDK inhibitor expression (P > .05). In the DAY1 group, CA4 decreased intracellular levels of cyclins D2, E, and A (P < .05). Despite recovery effects in the DAY5 group (increase of cyclins D1, D2, and A compared with DAY1 group; P < .05), the upregulation of the CDK inhibitor p21, increased D cyclins, and decreased cyclins E and A (P < .05) are compatible with a G1 arrest. CONCLUSIONS CA4 is a stronger inhibitor of the SMC cycle than sirolimus or paclitaxel and may represent an alternative for drug-eluting stents in atherosclerotic luminal stenosis. The effect of CA4 on neointima formation should be evaluated further.
Collapse
Affiliation(s)
- Daniel Spira
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Im Neuenheimer Feld 110, Heidelberg 69120, Germany.
| | - Gerd Grözinger
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls-University, Tübingen, Germany
| | - Nicole Domschke
- Department of Internal Medicine, SRH Hospital Sigmaringen, Sigmaringen, Germany
| | - Rüdiger Bantleon
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls-University, Tübingen, Germany
| | - Jörg Schmehl
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls-University, Tübingen, Germany
| | - Jakub Wiskirchen
- Department of Radiology and Nuclear Medicine, Franziskus Hospital, Bielefeld, Germany
| | - Benjamin Wiesinger
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls-University, Tübingen, Germany
| |
Collapse
|
34
|
Abstract
BACKGROUND The anti-malarial drug chloroquine has recently been discovered as a novel anti-tumor agent. This article is to review the recent development of chloroquine being used in cancer therapy. METHODS PubMed, ScienceDirect and ClinicalKey served as the major databases. Key words included 'chloroquine', 'cancer', and 'autophagy'. The publication date was up to June 2015. RESULTS Chloroquine mainly executes its anti-tumor function through inhibition of autophagy. It can accumulate inside the lysosome resulting in lysosomal membrane permeabilization (LMP) which will eventually lead to apoptosis. Chloroquine has been shown to stabilize p53 and induce p53-dependent apoptosis or cell cycle arrest. It can also inhibit ABC (ATP-binding cassette) family protein. The anti-cancer effect of chloroquine has been observed both in vitro and in vivo. However, it is considered more as a potential chemotherapy and radiotherapy sensitizer rather than an antineoplastic. CONCLUSION Although the utility of chloroquine is promising in cancer therapy, some safety issues have been brought to attention, and further studies on safety profile and the signs of clinical activity of chloroquine including its derivatives should be conducted.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pharmacy, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital , Chengdu , China
| | | | | | | |
Collapse
|
35
|
Magnetic nanosphere-guided site-specific delivery of vascular endothelial growth factor gene attenuates restenosis in rabbit balloon-injured artery. J Vasc Surg 2015; 63:226-33.e1. [PMID: 25595406 DOI: 10.1016/j.jvs.2014.11.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 11/15/2014] [Indexed: 11/21/2022]
Abstract
OBJECTIVE New and efficient strategies to protect endothelium or to enhance endothelial regrowth are important for treatment of restenosis after percutaneous transluminal angioplasty. Magnetic DNA microspheres are used to accelerate vascular endothelial growth factor (VEGF) re-endothelialization and to attenuate intimal hyperplasia in balloon-injured artery. This study aimed to assess DNA-gelatin magnetic nanospheres containing VEGF expression plasmids in vascular restenosis attenuation. METHODS Ninety-six rabbits underwent balloon injury and were randomly divided for gene transfer with naked VEGF plasmids (NAK group), magnetic VEGF microspheres (MIC group), and LacZ (CON group: naked LacZ plasmid and LacZ nanosphere subgroups). Serum and tissue VEGF levels were measured. Also, the ratios of intima area to media area were determined to assess neointima formation. RESULTS Microsphere gene delivery through the artery by a magnet resulted in VEGF overexpression in transfected arterial segments. Tissue VEGF integral optical densities were significantly increased in MIC rabbits compared with NAK animals. Serum VEGF was below detection in all animals. X-Gal staining showed higher transfection efficiency in the CON group. The impact of neointimal thickening was evaluated by light microscopy as the ratio of intima area to media area in cross sections. Significant differences in the ratio of intima area to media area were obtained between the NAK group (0.12 ± 0.02, 0.41 ± 0.03, 0.61 ± 0.05, and 0.72 ± 0.04 at 1, 2, 3, and 4 weeks, respectively) and the MIC group (0.06 ± 0.03, 0.20 ± 0.05, 0.25 ± 0.04, and 0.26 ± 0.03 at 1, 2, 3, and 4 weeks, respectively) at 2, 3, and 4 weeks (P < .05). CONCLUSIONS Intra-arterial VEGF gene delivery by magnetic microspheres significantly increased DNA stability, transfection efficiency, and targeting specificity, resulting in exogenous VEGF overexpression and attenuated intimal hyperplasia in balloon-injured artery.
Collapse
|
36
|
Abstract
Cardiovascular disease is the leading cause of death worldwide. As such, there is great interest in identifying novel mechanisms that govern the cardiovascular response to disease-related stress. First described in failing hearts, autophagy within the cardiovascular system has been widely characterized in cardiomyocytes, cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, and macrophages. In all cases, a window of optimal autophagic activity appears to be critical to the maintenance of cardiovascular homeostasis and function; excessive or insufficient levels of autophagic flux can each contribute to heart disease pathogenesis. In this Review, we discuss the potential for targeting autophagy therapeutically and our vision for where this exciting biology may lead in the future.
Collapse
|
37
|
Miyake T, Ihara S, Miyake T, Tsukada Y, Watanabe H, Matsuda H, Kiguchi H, Tsujimoto H, Nakagami H, Morishita R. Prevention of neointimal formation after angioplasty using nuclear factor-κB decoy oligodeoxynucleotide-coated balloon catheter in rabbit model. Circ Cardiovasc Interv 2014; 7:787-96. [PMID: 25406205 DOI: 10.1161/circinterventions.114.001522] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Despite the advent of drug-eluting stents, restenosis after endovascular intervention is still a major limitation in the treatment of cardiovascular disease. To regulate the multiple biological mechanisms underlying restenosis, we focused on inhibition of an important transcription factor, nuclear factor-kappaB (NFκB), using a decoy strategy. METHODS AND RESULTS For site-specific application of NFκB decoy oligodeoxynucleotides into target vessels during angioplasty, we developed a balloon catheter-based delivery system combined with biocompatible nanoparticles as oligodeoxynucleotides carriers. To clarify the therapeutic effect at the site of neointima, balloon angioplasty of the rabbit carotid arteries was performed at 4 weeks after initial endothelial denudation. This delivery system exhibited successful transfer of fluorescence-labeled nanospheres into the neointima in short-term contact with target vessels, and fluorescence could be detected ≥1 week after angioplasty. Consistently, local application of NFκB decoy oligodeoxynucleotides -loaded nanospheres resulted in significant inhibition of neointimal formation, associated with inhibition of NFκB binding activity in the injured arteries. The therapeutic effects were caused by inhibition of macrophage recruitment through the suppression of monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, and CC chemokine ligand 4 expression and inhibition of vascular smooth muscle cell growth via a decrease in the expression of cyclin A and proliferating cell nuclear antigen. Importantly, application of NFκB nanospheres accelerated restoration of the endothelial cell monolayer, associated with enhanced expression of phosphorylated Bcl-2 in endothelial cells. CONCLUSIONS A drug-coated balloon catheter using NFκB decoy oligodeoxynucleotides significantly inhibited the development of neointimal hyperplasia in rabbits. The present study indicates the possibility of a novel therapeutic option to prevent restenosis after angioplasty.
Collapse
Affiliation(s)
- Takashi Miyake
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Shinya Ihara
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Tetsuo Miyake
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Yusuke Tsukada
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Hajime Watanabe
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Hiroaki Matsuda
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Hideki Kiguchi
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Hiroyuki Tsujimoto
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Hironori Nakagami
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.)
| | - Ryuichi Morishita
- From the Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan (T.M., T.M., R.M.); Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Osaka, Japan (T.M., H.N.); AnGes MG, Inc, Osaka, Japan (S.I., H.W.); Pharmaceutical and Beauty Science Research Center, Hosokawa Micron Corporation, Osaka, Japan (Y.T., H.T.); and Togo Medikit Co, Ltd, Miyazaki, Japan (H.M., H.K.).
| |
Collapse
|
38
|
Ye LX, Yu J, Liang YX, Zeng JS, Huang RX, Liao SJ. Beclin 1 knockdown retards re-endothelialization and exacerbates neointimal formation via a crosstalk between autophagy and apoptosis. Atherosclerosis 2014; 237:146-54. [DOI: 10.1016/j.atherosclerosis.2014.08.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/13/2014] [Accepted: 08/28/2014] [Indexed: 01/15/2023]
|
39
|
Roubille F, Busseuil D, Shi Y, Nachar W, Mihalache-Avram T, Mecteau M, Gillis MA, Brand G, Théberge-Julien G, Brodeur MR, Kernaleguen AE, Gombos M, Rhéaume E, Tardif JC. The interleukin-1β modulator gevokizumab reduces neointimal proliferation and improves reendothelialization in a rat carotid denudation model. Atherosclerosis 2014; 236:277-85. [DOI: 10.1016/j.atherosclerosis.2014.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 06/20/2014] [Accepted: 07/11/2014] [Indexed: 01/11/2023]
|
40
|
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide, despite the significant advances in medicine. Autophagy, a process of self-cannibalization employed by mammalian cells for the recycling of cellular contents, is altered not only in a number of CVDs, but in other diseases, as well. Many FDA-approved drugs are known to induce autophagy-mediated side effects in the cardiovascular system. In some cases, such drug-induced autophagy could be harnessed and used for treating CVD, greatly reducing the duration and cost of CVD treatments. However, because the induction of autophagy in cardiovascular targets can be both adaptive and maladaptive under specific settings, the challenge is to determine whether the changes stimulated by drug-induced autophagy are, in fact, beneficial. In this review, we surveyed a number of CVDs in which autophagy is known to occur, and we also address the role of FDA-approved drugs for which autophagy-mediated side effects occur within the cardiovascular system. The therapeutic potential of using small molecule modulators of autophagy in the management of CVD progression is discussed.
Collapse
|
41
|
Gabriel D, Cohen-Karni T, Huang D, Chiang HH, Kohane DS. Photoactive electrospun fibers for inducing cell death. Adv Healthc Mater 2014; 3:494-9. [PMID: 24574265 DOI: 10.1002/adhm.201300318] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/16/2013] [Indexed: 11/07/2022]
Abstract
A photoactive electrospun material producing reactive oxygen species (ROS) upon light irradiation is reported. The phototoxicity of the generated ROS is spatially restricted to the fiber-tissue interface by conjugation of the photosensitizer to a macromolecule. Photo-triggered ROS is produced on demand and repeatedly. It induces death of mammalian cells growing on the material surface with high spatial resolution.
Collapse
Affiliation(s)
- Doris Gabriel
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine; Children's Hospital Boston, Harvard Medical School; 300 Longwood Avenue Boston MA 02115 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Tzahi Cohen-Karni
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine; Children's Hospital Boston, Harvard Medical School; 300 Longwood Avenue Boston MA 02115 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - David Huang
- Department of Chemical Engineering; Massachusetts Institute of Technology; 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Homer H. Chiang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine; Children's Hospital Boston, Harvard Medical School; 300 Longwood Avenue Boston MA 02115 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine; Children's Hospital Boston, Harvard Medical School; 300 Longwood Avenue Boston MA 02115 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; 77 Massachusetts Avenue Cambridge MA 02139 USA
| |
Collapse
|
42
|
Jiang P, Lan Y, Luo J, Ren YL, Liu DG, Pang JX, Liu J, Li J, Wang C, Cai JP. Rapamycin promoted thrombosis and platelet adhesion to endothelial cells by inducing membrane remodeling. BMC Cell Biol 2014; 15:7. [PMID: 24564184 PMCID: PMC3936831 DOI: 10.1186/1471-2121-15-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 02/06/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recently, evidence indicated that the rapamycin-eluting stent which was used worldwide may contribute to an increased risk for thrombosis. On the contrary, other researchers found it was safe. Thus, it is necessary to clarify the effect of rapamycin on thrombosis and the corresponding mechanisms. RESULTS The effects of rapamycin in vivo were evaluated by modified deep vein thrombosis animal model. The platelets were from healthy volunteers and the platelet-endothelium (purchased from ATCC) adhesion in cultured endothelial cells was assessed. Membrane rufflings in endothelial cells were examined by confocal and electron microscope. Thrombus formation increased in rats that were injected with rapamycin. Electron microscope analysis exhibited microvilli on the rapamycin-treated endothelium in rats. Rapamycin enhanced membrane ruffling in human umbilical vein endothelial cells (HUVECs) and adhesion of platelets to HUVECs. The platelet-HUVECs adhesion was attenuated when cells were treated with cytochalacin B. Inhibition of autophagy by 3-methyladenine led to suppression of membrane ruffles in HUVECs and augmentation of platelet-endothelial adhesion. CONCLUSIONS In conclusion, we found that endothelial membrane remodeling induced by rapamycin is crucial for the adhesion of platelets to endothelial cells and thereby for thrombosis in vivo, and that the endothelial membrane remodeling is autophagy dependent.
Collapse
Affiliation(s)
- Ping Jiang
- The Key Laboratory of Geriatrics, Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, No,1, DaHua Road, Dong Dan, Beijing 100730, P,R,China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Martinet W, De Loof H, De Meyer GRY. mTOR inhibition: a promising strategy for stabilization of atherosclerotic plaques. Atherosclerosis 2014; 233:601-607. [PMID: 24534455 DOI: 10.1016/j.atherosclerosis.2014.01.040] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/10/2014] [Accepted: 01/19/2014] [Indexed: 01/06/2023]
Abstract
Statins are currently able to stabilize atherosclerotic plaques by lowering plasma cholesterol and pleiotropic effects, but a residual risk for atherosclerotic disease remains. Therefore, effective prevention of atherosclerosis and treatment of its complications is still a major clinical challenge. A large body of evidence indicates that mammalian target of rapamycin (mTOR) inhibitors such as rapamycin or everolimus have pleiotropic anti-atherosclerotic effects so that these drugs can be used as add-on therapy to prevent or delay the pathogenesis of atherosclerosis. Moreover, bioresorbable scaffolds eluting everolimus trigger a healing process in the vessel wall, both in pigs and humans, that results in late lumen enlargement and plaque regression. At present, this phenomenon of atheroregression is poorly understood. However, given that mTOR inhibitors suppress cell proliferation and trigger autophagy, a cellular survival pathway and a process linked to cholesterol efflux, we hypothesize that these compounds can inhibit (or reverse) the basic mechanisms that control plaque growth and destabilization. Unfortunately, adverse effects associated with mTOR inhibitors such as dyslipidemia and hyperglycemia have recently been identified. Dyslipidemia is manageable via statin treatment, while the anti-diabetic drug metformin would prevent hyperglycemia. Because metformin has beneficial macrovascular effects, this drug in combination with an mTOR inhibitor might have significant promise to treat patients with unstable plaques. Moreover, both statins and metformin are known to inhibit mTOR via AMPK activation so that they would fully exploit the beneficial effects of mTOR inhibition in atherosclerosis.
Collapse
Affiliation(s)
- Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Hans De Loof
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| |
Collapse
|
44
|
Yurdagul A, Kleinedler JJ, McInnis MC, Khandelwal AR, Spence AL, Orr AW, Dugas TR. Resveratrol promotes endothelial cell wound healing under laminar shear stress through an estrogen receptor-α-dependent pathway. Am J Physiol Heart Circ Physiol 2014; 306:H797-806. [PMID: 24464753 DOI: 10.1152/ajpheart.00892.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Restenosis is an adverse outcome of angioplasty, characterized by vascular smooth muscle cell (VSMC) hyperplasia. However, therapies targeting VSMC proliferation delay re-endothelialization, increasing the risk of thrombosis. Resveratrol (RESV) inhibits restenosis and promotes re-endothelialization after arterial injury, but in vitro studies assessing RESV-mediated effects on endothelial cell growth contradict these findings. We thus hypothesized that fluid shear stress, mimicking physiological blood flow, would recapitulate RESV-dependent endothelial cell wound healing. Since RESV is an estrogen receptor (ER) agonist, we tested whether RESV promotes re-endothelialization through an ER-α-dependent mechanism. Mice fed a high-fat diet or a diet supplemented with RESV were subjected to carotid artery injury. At 7 days after injury, RESV significantly accelerated re-endothelialization compared with vehicle. In vitro wound healing assays demonstrated that RESV exhibits cell-type selectivity, inhibiting VSMC, but not endothelial cell growth. Under laminar shear stress (LSS), RESV dramatically enhanced endothelial cell wound healing and increased both the activation of extracellular signal-regulated kinase (ERK) and endothelial cell proliferation. Under LSS, small interfering RNA against ER-α, but not endothelial nitric oxide synthase, abolished RESV-induced ERK activation, endothelial cell proliferation, and wound healing. Thus these studies suggest that the EC phenotype induced by LSS better models the prohealing effects of RESV and that RESV and LSS interact to promote an ER-α-dependent mitogenic effect in endothelial cells.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | | | | | | | | | | | | |
Collapse
|
45
|
Urbanek T, Kuczmik W, Basta-Kaim A, Gabryel B. Rapamycin induces of protective autophagy in vascular endothelial cells exposed to oxygen-glucose deprivation. Brain Res 2014; 1553:1-11. [PMID: 24462935 DOI: 10.1016/j.brainres.2014.01.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 01/14/2014] [Accepted: 01/15/2014] [Indexed: 02/06/2023]
Abstract
The protective potential of rapamycin has been reported in a few experimental models of brain ischemia, both in vivo and in vitro. Although the precise cellular processes underlying the neuroprotective effects of rapamycin in experimental models of stroke remain unknown, the current experimental data suggest that the mechanism of action of the drug may result from the mTOR-mediated autophagy induction. However, it is unclear whether the activation of autophagy acts as a pro-death or pro-survival factor in vascular endothelial cells in ischemic brain damage. It seems to be very important, since stroke affects not only neurons and astrocytes but also microvessels. In the present study, we used human umbilical vein endothelial cells (HUVEC) subjected to ischemia-simulating conditions (combined oxygen and glucose deprivation, OGD) for 6h to determine potential effect of rapamycin-induced autophagy on HUVEC damage. The drug at concentrations of 100 and 1000nM increased the expression of Beclin 1 and LC3-II together with a significant increase in the p62 degradation in ischemic HUVEC. Treatment with rapamycin in OGD significantly increased the cell viability, indicating that the drug exerts cytoprotective effect. The inhibition of Beclin 1 by siRNAs significantly attenuated the expression of autophagy-related proteins and reduced HUVEC viability following OGD and rapamycin treatment. Our findings demonstrated that toxicity of simulated ischemia conditions were enhanced in HUVEC when autophagy was blocked, and that rapamycin effectively prevented OGD-evoked damage by induction of protective autophagy via inhibition of the mTOR pathway.
Collapse
Affiliation(s)
- Tomasz Urbanek
- Department of General and Vascular Surgery, Medical University of Silesia, Ziołowa 45/47, PL 40-635 Katowice, Poland
| | - Wacław Kuczmik
- Department of General and Vascular Surgery, Medical University of Silesia, Ziołowa 45/47, PL 40-635 Katowice, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343 Kraków, Poland
| | - Bożena Gabryel
- Department of Pharmacology, Medical University of Silesia, Medyków 18, PL 40-752 Katowice, Poland.
| |
Collapse
|
46
|
Liu Y, Wang X, Lu J. Dynamic observation of artery endothelial cell layers in response to bare metal stent and paclitaxel-eluting stent in vitro. J Interv Cardiol 2014; 27:182-90. [PMID: 24433433 DOI: 10.1111/joic.12092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES We aimed to investigate the mechanism of paclitaxel-eluting stent (PES) induced thrombosis by real-time dynamic monitoring of live endothelial cells. BACKGROUND PES is widely used in clinics to inhibit restenosis effectively. However, late stent thrombosis is a major concern with the use of PES. METHODS We established an in vitro cell culture system to mimic the close contact of endothelial cells with PES struts. The dynamic response of porcine innominate artery endothelial cells (PIECs) to stents was observed using a bright field microscope and a high-resolution charge-coupled device RESULTS PES changed elongated PIECs to round PIECs within 24 hours. Paracellular gaps were readily observed in a PIEC monolayer exposed to PES. By contrast, paracellular gaps were almost undetectable in an endothelial monolayer incubated with bare metal stent (BMS). As incubation time was prolonged (days 5-9), round PIECs returned to their elongated shape, but paracellular gaps were retained at lower frequencies and smaller sizes than those on days 1 and 2. In addition, the PIEC monolayer in the PES group retained an uneven surface topology during incubation, whereas PIECs in the BMS group developed a smooth surface of epithelial cell sheets on days 5-7. CONCLUSION Our findings showed that the shift of cell shape causes impaired integrity of the monolayer characteristic with enlarged paracellular gaps and uneven surface topology in exposure to PES. The results might serve as structural information for understanding the mechanism of PES-induced early and late thrombosis.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Cardiology, Dongzhimen Hospital, First Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | | | | |
Collapse
|
47
|
Survival or death: disequilibrating the oncogenic and tumor suppressive autophagy in cancer. Cell Death Dis 2013; 4:e892. [PMID: 24176850 PMCID: PMC3920945 DOI: 10.1038/cddis.2013.422] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 09/20/2013] [Accepted: 09/23/2013] [Indexed: 02/05/2023]
Abstract
Autophagy (macroautophagy) is an evolutionarily conserved lysosomal degradation process, in which a cell degrades long-lived proteins and damaged organelles. Recently, accumulating evidence has revealed the core molecular machinery of autophagy in carcinogenesis; however, the intricate relationship between autophagy and cancer continue to remain an enigma. Why does autophagy have either pro-survival (oncogenic) or pro-death (tumor suppressive) role at different cancer stages, including cancer stem cell, initiation and progression, invasion and metastasis, as well as dormancy? How does autophagy modulate a series of oncogenic and/or tumor suppressive pathways, implicated in microRNA (miRNA) involvement? Whether would targeting the oncogenic and tumor suppressive autophagic network be a novel strategy for drug discovery? To address these problems, we focus on summarizing the dynamic oncogenic and tumor suppressive roles of autophagy and their relevant small-molecule drugs, which would provide a new clue to elucidate the oncosuppressive (survival or death) autophagic network as a potential therapeutic target.
Collapse
|
48
|
Zhang X, Chen L, Ouyang L, Cheng Y, Liu B. Plant natural compounds: targeting pathways of autophagy as anti-cancer therapeutic agents. Cell Prolif 2012; 45:466-76. [PMID: 22765290 PMCID: PMC6496896 DOI: 10.1111/j.1365-2184.2012.00833.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/04/2012] [Indexed: 12/16/2022] Open
Abstract
Natural compounds derived from plant sources are well characterized as possessing a wide variety of remarkable anti-tumour properties, for example modulating programmed cell death, primarily referring to apoptosis, and autophagy. Distinct from apoptosis, autophagy (an evolutionarily conserved, multi-step lysosomal degradation process in which a cell destroys long-lived proteins and damaged organelles) may play crucial regulatory roles in many pathological processes, most notably in cancer. In this review, we focus on highlighting several representative plant natural compounds such as curcumin, resveratrol, paclitaxel, oridonin, quercetin and plant lectin - that may lead to cancer cell death - for regulation of some core autophagic pathways, involved in Ras-Raf signalling, Beclin-1 interactome, BCR-ABL, PI3KCI/Akt/mTOR, FOXO1 signalling and p53. Taken together, these findings would provide a new perspective for exploiting more plant natural compounds as potential novel anti-tumour drugs, by targeting the pathways of autophagy, for future cancer therapeutics.
Collapse
Affiliation(s)
- X. Zhang
- Department of Natural Products ChemistrySchool of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangChina
| | - L.‐X. Chen
- Department of Natural Products ChemistrySchool of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyangChina
| | - L. Ouyang
- The State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Y. Cheng
- Department of Pharmacology and The Penn State Cancer InstituteThe Pennsylvania State University College of MedicineMilton S. Hershey Medical CenterHersheyPennsylvaniaUSA
| | - B. Liu
- The State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
49
|
Douglas G, Van Kampen E, Hale AB, McNeill E, Patel J, Crabtree MJ, Ali Z, Hoerr RA, Alp NJ, Channon KM. Endothelial cell repopulation after stenting determines in-stent neointima formation: effects of bare-metal vs. drug-eluting stents and genetic endothelial cell modification. Eur Heart J 2012; 34:3378-88. [PMID: 23008511 PMCID: PMC3827553 DOI: 10.1093/eurheartj/ehs240] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Aims Understanding endothelial cell repopulation post-stenting and how this modulates in-stent restenosis is critical to improving arterial healing post-stenting. We used a novel murine stent model to investigate endothelial cell repopulation post-stenting, comparing the response of drug-eluting stents with a primary genetic modification to improve endothelial cell function. Methods and results Endothelial cell repopulation was assessed en face in stented arteries in ApoE−/− mice with endothelial-specific LacZ expression. Stent deployment resulted in near-complete denudation of endothelium, but was followed by endothelial cell repopulation, by cells originating from both bone marrow-derived endothelial progenitor cells and from the adjacent vasculature. Paclitaxel-eluting stents reduced neointima formation (0.423 ± 0.065 vs. 0.240 ± 0.040 mm2, P = 0.038), but decreased endothelial cell repopulation (238 ± 17 vs. 154 ± 22 nuclei/mm2, P = 0.018), despite complete strut coverage. To test the effects of selectively improving endothelial cell function, we used transgenic mice with endothelial-specific overexpression of GTP-cyclohydrolase 1 (GCH-Tg) as a model of enhanced endothelial cell function and increased NO production. GCH-Tg ApoE−/− mice had less neointima formation compared with ApoE−/− littermates (0.52 ± 0.08 vs. 0.26 ± 0.09 mm2, P = 0.039). In contrast to paclitaxel-eluting stents, reduced neointima formation in GCH-Tg mice was accompanied by increased endothelial cell coverage (156 ± 17 vs. 209 ± 23 nuclei/mm2, P = 0.043). Conclusion Drug-eluting stents reduce not only neointima formation but also endothelial cell repopulation, independent of strut coverage. In contrast, selective targeting of endothelial cell function is sufficient to improve endothelial cell repopulation and reduce neointima formation. Targeting endothelial cell function is a rational therapeutic strategy to improve vascular healing and decrease neointima formation after stenting.
Collapse
Affiliation(s)
- Gillian Douglas
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ramakrishnan R, Huang C, Cho HI, Lloyd M, Johnson J, Ren X, Altiok S, Sullivan D, Weber J, Celis E, Gabrilovich DI. Autophagy induced by conventional chemotherapy mediates tumor cell sensitivity to immunotherapy. Cancer Res 2012; 72:5483-93. [PMID: 22942258 DOI: 10.1158/0008-5472.can-12-2236] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Autophagy attenuates the efficacy of conventional chemotherapy but its effects on immunotherapy have been little studied. Here, we report that chemotherapy renders tumor cells more susceptible to lysis by CTL in vivo. Moreover, bystander tumor cells that did not express antigen were killed by CTL. This effect was mediated by transient but dramatic upregulation of the mannose-6-phosphate receptor (MPR) on the tumor cell surface. Antitumor effects of combined treatment related to the kinetics of MPR upregulation and abrogation of this event abolished the combined effect of immunotherapy and chemotherapy. MPR accumulation on the tumor cell surface during chemotherapy was observed in different mouse tumor models and in patients with multiple myeloma. Notably, this effect was the result of redistribution of the receptor caused by chemotherapy-inducible autophagy. Together, our findings reveal one molecular mechanism through which the antitumor effects of conventional cancer chemotherapy and immunotherapy are realized.
Collapse
Affiliation(s)
- Rupal Ramakrishnan
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|