1
|
Chao CL, Applewhite B, Reddy NK, Matiuto N, Dang C, Jiang B. Advances and challenges in regenerative therapies for abdominal aortic aneurysm. Front Cardiovasc Med 2024; 11:1369785. [PMID: 38895536 PMCID: PMC11183335 DOI: 10.3389/fcvm.2024.1369785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a significant source of mortality worldwide and carries a mortality of greater than 80% after rupture. Despite extensive efforts to develop pharmacological treatments, there is currently no effective agent to prevent aneurysm growth and rupture. Current treatment paradigms only rely on the identification and surveillance of small aneurysms, prior to ultimate open surgical or endovascular repair. Recently, regenerative therapies have emerged as promising avenues to address the degenerative changes observed in AAA. This review briefly outlines current clinical management principles, characteristics, and pharmaceutical targets of AAA. Subsequently, a thorough discussion of regenerative approaches is provided. These include cellular approaches (vascular smooth muscle cells, endothelial cells, and mesenchymal stem cells) as well as the delivery of therapeutic molecules, gene therapies, and regenerative biomaterials. Lastly, additional barriers and considerations for clinical translation are provided. In conclusion, regenerative approaches hold significant promise for in situ reversal of tissue damages in AAA, necessitating sustained research and innovation to achieve successful and translatable therapies in a new era in AAA management.
Collapse
Affiliation(s)
- Calvin L. Chao
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Brandon Applewhite
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Chicago, IL, United States
| | - Nidhi K. Reddy
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Natalia Matiuto
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Caitlyn Dang
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Bin Jiang
- Division of Vascular Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Chicago, IL, United States
| |
Collapse
|
2
|
Applewhite B, Andreopoulos F, Vazquez-Padron RI. Periadventitial biomaterials to improve arteriovenous fistula and graft outcomes. J Vasc Access 2024; 25:713-727. [PMID: 36349745 DOI: 10.1177/11297298221135621] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
Periadventitial biomaterials have been employed for nearly three decades to promote adaptive venous remodeling following hemodialysis vascular access creation in preclinical models and clinical trials. These systems are predicated on the combination of scaffolds, hydrogels, and/or particles with therapeutics (small molecules, proteins, genes, and cells) to prevent venous stenosis and subsequent maturation failure. Periadventitial biomaterial therapies have evolved from simple drug delivery vehicles for traditional drugs to more thoughtful designs tailored to the pathophysiology of access failure. The emergence of tissue engineering strategies and gene therapies are another exciting new direction. Despite favorable results in experimental and preclinical studies, no periadventitial therapy has been clinically approved to improve vascular access outcomes. After conducting an exhaustive review of the literature, we identify the seminal studies and clinical trials that utilize periadventitial biomaterials and discuss the key features of each biomaterial format and their respective shortcomings as they pertain to access maturation. This review provides a foundation from which clinicians, surgeons, biologists, and engineers can refer to and will hopefully inspire thoughtful, translatable treatments to finally address access failure.
Collapse
Affiliation(s)
- Brandon Applewhite
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami College of Engineering, Coral Gables, FL, USA
| | - Fotios Andreopoulos
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami College of Engineering, Coral Gables, FL, USA
| | - Roberto I Vazquez-Padron
- Department of Biomedical Engineering, University of Miami College of Engineering, Coral Gables, FL, USA
| |
Collapse
|
3
|
Fernandes A, Miéville A, Grob F, Yamashita T, Mehl J, Hosseini V, Emmert MY, Falk V, Vogel V. Endothelial-Smooth Muscle Cell Interactions in a Shear-Exposed Intimal Hyperplasia on-a-Dish Model to Evaluate Therapeutic Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202317. [PMID: 35971167 PMCID: PMC9534971 DOI: 10.1002/advs.202202317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Indexed: 05/25/2023]
Abstract
Intimal hyperplasia (IH) represents a major challenge following cardiovascular interventions. While mechanisms are poorly understood, the inefficient preventive methods incentivize the search for novel therapies. A vessel-on-a-dish platform is presented, consisting of direct-contact cocultures with human primary endothelial cells (ECs) and smooth muscle cells (SMCs) exposed to both laminar pulsatile and disturbed flow on an orbital shaker. With contractile SMCs sitting below a confluent EC layer, a model that successfully replicates the architecture of a quiescent vessel wall is created. In the novel IH model, ECs are seeded on synthetic SMCs at low density, mimicking reendothelization after vascular injury. Over 3 days of coculture, ECs transition from a network conformation to confluent 2D islands, as promoted by pulsatile flow, resulting in a "defected" EC monolayer. In defected regions, SMCs incorporated plasma fibronectin into fibers, increased proliferation, and formed multilayers, similarly to IH in vivo. These phenomena are inhibited under confluent EC layers, supporting therapeutic approaches that focus on endothelial regeneration rather than inhibiting proliferation, as illustrated in a proof-of-concept experiment with Paclitaxel. Thus, this in vitro system offers a new tool to study EC-SMC communication in IH pathophysiology, while providing an easy-to-use translational disease model platform for low-cost and high-content therapeutic development.
Collapse
Affiliation(s)
- Andreia Fernandes
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Arnaud Miéville
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Franziska Grob
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Tadahiro Yamashita
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
- Present address:
Department of System Design EngineeringKeio University108‐8345YokohamaJapan
| | - Julia Mehl
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
- Present address:
Julius Wolff InstituteBerlin Institute of HealthCharité Universitätsmedizin Berlin10117BerlinGermany
| | - Vahid Hosseini
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Maximilian Y. Emmert
- Department of Cardiovascular SurgeryCharité Universitätsmedizin Berlin10117BerlinGermany
- Department of Cardiothoracic and Vascular SurgeryGerman Heart Center Berlin13353BerlinGermany
- Institute for Regenerative Medicine (IREM)University of Zurich8006ZurichSwitzerland
| | - Volkmar Falk
- Department of Cardiovascular SurgeryCharité Universitätsmedizin Berlin10117BerlinGermany
- Department of Cardiothoracic and Vascular SurgeryGerman Heart Center Berlin13353BerlinGermany
- Department of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Viola Vogel
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| |
Collapse
|
4
|
Parikh SA, Edelman ER. Tissue-Engineered Endothelial Cells Induce Sustained Vascular Healing Through Early Induction of Vascular Repair. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Abstract
Background
Perivascular implantation of tissue-engineered endothelial cells (TEEC) after vascular injury profoundly inhibits neointimal hyperplasia. However, the time course and mechanism by which this effect occurs remain unknown. By developing genetically modified TEEC that express a “suicide gene,” we can control the time during which the TEEC could exert their effect and determine the length of time TEEC need to be present following vascular injury to exert their inhibitory effect on long-term neointimal hyperplasia.
Methods
Bovine aortic endothelial cells (BAE) were transfected with the human herpes simplex virus thymidine kinase (tk) gene to render them sensitive to ganciclovir (GCV). These BAE+tk were grown to confluence on Gelfoam and shown to have the same growth kinetics and biologic potency as control cells but were sensitive to GCV at low concentrations. The BAE+tk were grown on Gelfoam and placed in the perivascular space around balloon-injured rat carotid arteries. Rats randomly received BAE-tk, BAE+tk, or nothing (control) after balloon injury. GCV was administered early (days 1–7), late (days 5–11), or not at all.
Results
Two weeks after injury, extensive neointimal hyperplasia was observed in control animals with an intima:media (I:M) area ratio of 0.80 ± 0.19. Early administration of GCV killed the BAE in constructs with TK sensitivity and eliminated the impact of TEEC regulation of intimal hyperplasia (0.45 ± 0.06). Intimal hyperplasia was still effectively reduced in animals with implants containing BAE-tk or BAE+tk which received GCV late (0.11 ± 0.04 and 0.19 ± 0.05). Immunohistochemistry demonstrated the lethal effect of GCV on TK-sensitive cells.
Conclusions
The application of perivascular TEEC for only the first few days after injury had a significant inhibitory effect on intimal hyperplasia. This is in contrast to the results obtained in this same animal model with the infusion of isolated anti-smooth muscle cell proliferative agents. This suggests that the mechanism of action of TEEC may be upstream from smooth muscle cell proliferation. Moreover, the use of this technique to further elucidate biologic mechanisms will prove invaluable in the tissue engineering field.
Lay Summary
We report a novel, genetically altered tissue-engineered endothelial cell (TEEC) implant that inhibits neointimal hyperplasia after experimental vascular injury. The viability of these implants can be carefully controlled and suggest a putative mechanism by which TEEC recapitulate control over the vascular response to injury.
Collapse
|
5
|
Gooch KJ, Firstenberg MS, Shrefler BS, Scandling BW. Biomechanics and Mechanobiology of Saphenous Vein Grafts. J Biomech Eng 2019; 140:2666246. [PMID: 29222565 DOI: 10.1115/1.4038705] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Indexed: 11/08/2022]
Abstract
Within several weeks of use as coronary artery bypass grafts (CABG), saphenous veins (SV) exhibit significant intimal hyperplasia (IH). IH predisposes vessels to thrombosis and atherosclerosis, the two major modes of vein graft failure. The fact that SV do not develop significant IH in their native venous environment coupled with the rapidity with which they develop IH following grafting into the arterial circulation suggests that factors associated with the isolation and preparation of SV and/or differences between the venous and arterial environments contribute to disease progression. There is strong evidence suggesting that mechanical trauma associated with traditional techniques of SV preparation can significantly damage the vessel and might potentially reduce graft patency though modern surgical techniques reduces these injuries. In contrast, it seems possible that modern surgical technique, specifically endoscopic vein harvest, might introduce other mechanical trauma that could subtly injure the vein and perhaps contribute to the reduced patency observed in veins harvested using endoscopic techniques. Aspects of the arterial mechanical environment influence remodeling of SV grafted into the arterial circulation. Increased pressure likely leads to thickening of the medial wall but its role in IH is less clear. Changes in fluid flow, including increased average wall shear stress, may reduce IH while disturbed flow likely increase IH. Nonmechanical stimuli, such as exposure to arterial levels of oxygen, may also have a significant but not widely recognized role in IH. Several potentially promising approaches to alter the mechanical environment to improve graft patency are including extravascular supports or altered graft geometries are covered.
Collapse
Affiliation(s)
- Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University, 290 Bevis Hall 1080 Carmack Drive, Columbus, OH 43210.,Davis Heart Lung Research Institute, The Ohio State University, Columbus, OH 43210 e-mail:
| | - Michael S Firstenberg
- Surgery and Integrative Medicine, Northeast Ohio Medical Universities, Akron, OH 44309
| | - Brittany S Shrefler
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Benjamin W Scandling
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
6
|
Disruptive technological advances in vascular access for dialysis: an overview. Pediatr Nephrol 2018; 33:2221-2226. [PMID: 29188361 DOI: 10.1007/s00467-017-3853-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 12/31/2022]
Abstract
End-stage kidney disease (ESKD), one of the most prevalent diseases in the world and with increasing incidence, is associated with significant morbidity and mortality. Current available modes of renal replacement therapy (RRT) include dialysis and renal transplantation. Though renal transplantation is the preferred and ideal mode of RRT, this modality may not be available to all patients with ESKD. Moreover, renal transplant recipients are constantly at risk of complications associated with immunosuppression and immunosuppressant use, and posttransplant lymphoproliferative disorder. Dialysis may be the only available modality in certain patients. However, dialysis has its limitations, which include issues associated with lack of vascular access, risks of infections and vascular thrombosis, decreased quality of life, and absence of biosynthetic functions of the kidney. In particular, the creation and maintenance of hemodialysis vascular access in children poses a unique set of challenges to the pediatric nephrologist owing to the smaller vessel diameters and vascular hyperreactivity compared with adult patients. Vascular access issues continue to be one of the major limiting factors prohibiting the delivery of adequate dialysis in ESKD patients and is the Achilles' heel of hemodialysis. This review aims to provide a critical overview of disruptive technological advances and innovations for vascular access. Novel strategies in preventing neointimal hyperplasia, novel bioengineered products, grafts and devices for vascular access will be discussed. The potential impact of these solutions on improving the morbidity encountered by dialysis patients will also be examined.
Collapse
|
7
|
Lopez-Moya M, Melgar-Lesmes P, Kolandaivelu K, de la Torre Hernández JM, Edelman ER, Balcells M. Optimizing Glutaraldehyde-Fixed Tissue Heart Valves with Chondroitin Sulfate Hydrogel for Endothelialization and Shielding against Deterioration. Biomacromolecules 2018. [PMID: 29539266 DOI: 10.1021/acs.biomac.8b00077] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Porcine glutaraldehyde-fixed pericardium is widely used to replace human heart valves. Despite the stabilizing effects of glutaraldehyde fixation, the lack of endothelialization and the occurrence of immune reactions contribute to calcification and structural valve deterioration, which is particularly significant in young patients, in whom valve longevity is crucial. This report shows an optimization system with which to enhance endothelialization of fixed pericardium to mimic the biological function of a native heart valve. The glutaraldehyde detoxification, together with the application of a biodegradable methacrylated chondroitin sulfate hydrogel, reduces aldehydes cytotoxicity, increases the migration and proliferation of endothelial cells and the recruitment of endothelial cell progenitors, and confers thromboresistance in fixed pericardium. The combination of glutaraldehyde detoxification and a coating with chondroitin sulfate hydrogel promotes in situ mechanisms of endothelialization in fixed pericardium. We offer a new solution for improving the long life of bioprosthetic valves and exploring the means of making valves suitable to endothelialization.
Collapse
Affiliation(s)
- Mario Lopez-Moya
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science , Cambridge , Massachusetts 02139 , United States.,Bioengineering Department , Institut Químic de Sarrià, Ramon Llull Univ , Barcelona 08017 , Spain
| | - Pedro Melgar-Lesmes
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science , Cambridge , Massachusetts 02139 , United States.,Department of Biomedicine , Fundació Clínic per a la Recerca Biomèdica, University of Barcelona , Barcelona 08036 , Spain
| | - Kumaran Kolandaivelu
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science , Cambridge , Massachusetts 02139 , United States.,Cardiovascular Division , Brigham and Women's Hospital and Harvard Medical School , Boston , Massachusetts 02115 , United States
| | | | - Elazer R Edelman
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science , Cambridge , Massachusetts 02139 , United States.,Cardiovascular Division , Brigham and Women's Hospital and Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Mercedes Balcells
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science , Cambridge , Massachusetts 02139 , United States.,Bioengineering Department , Institut Químic de Sarrià, Ramon Llull Univ , Barcelona 08017 , Spain
| |
Collapse
|
8
|
Saemisch M, Nickmann M, Riesinger L, Edelman ER, Methe H. 3D matrix-embedding inhibits cycloheximide-mediated sensitization to TNF-alpha-induced apoptosis of human endothelial cells. J Tissue Eng Regen Med 2017; 12:1085-1096. [PMID: 29131527 DOI: 10.1002/term.2609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 10/23/2017] [Accepted: 10/28/2017] [Indexed: 12/30/2022]
Abstract
The programmed form of cell death (apoptosis) is essential for normal development of multicellular organisms. Dysregulation of apoptosis has been linked with embryonal death and is involved in the pathophysiology of various diseases. Others and we previously demonstrated endothelial biology being intertwined with biochemical and structural composition of the subendothelial basement membrane. We now demonstrate that a three-dimensional growing environment significantly shields endothelial cells from cytokine-induced apoptosis. Detailed analysis reveals differences in intracellular signaling pathways in naive endothelial cells and cytokine-stimulated endothelial cells when cells are grown within a three-dimensional collagen-based matrix compared to cells grown on two-dimensional tissue culture plates. Main findings are significantly reduced p53 expression and level of p38-phosphorylation in three-dimensional grown endothelial cells. Despite similar concentrations of focal adhesion kinase, three-dimensional matrix-embedded endothelial cells express significantly less tyrosine-phosphorylated focal adhesion kinase. Pretreatment with antibodies against integrin αv β3 partially reversed the protective effect of three-dimensional matrix-embedding on endothelial apoptosis. Our findings provide detailed insights into the mechanisms of endothelial apoptosis with respect to the spatial matrix environment. These results enhance our understanding of endothelial biology and may otherwise help in the design of tissue-engineered materials. Furthermore, findings on focal adhesion kinase phosphorylation might enhance our understanding of clinical studies with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Michael Saemisch
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany.,Department of Cardiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Lisa Riesinger
- Department of Cardiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Elazer R Edelman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heiko Methe
- Department of Cardiology, Ludwig-Maximilians-University Munich, Munich, Germany.,Kliniken an der Paar, Aichach, Germany.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
9
|
Paneghetti L, Ng YSE. A novel endothelial-derived anti-inflammatory activity significantly inhibits spontaneous choroidal neovascularisation in a mouse model. Vasc Cell 2016; 8:2. [PMID: 27175278 PMCID: PMC4864930 DOI: 10.1186/s13221-016-0036-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 05/03/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Endothelial cells (EC) grown on collagen particles inhibit intimal hyperplasia in animal models when applied perivascularly, and this effect appears to be, at least in part, the result of EC-derived soluble factors that suppress local vascular inflammation. To elucidate the molecular basis of the therapeutic effects of EC grown on collagen particles, the anti-inflammatory activity of conditioned medium from these cells was characterized. METHODS Human aortic EC (HAEC) and, for chromatin immunoprecipitation assays, human umbilical vein EC (HUVEC) were treated with tumor necrosis factor alpha (TNFα) in the presence of conditioned medium generated by HAEC grown on collagen particles (ECPCM), and the anti-inflammatory effects were evaluated by analysing the expression of the inflammation-related adhesion molecules E-selectin and vascular cell adhesion molecule-1 (VCAM-1). The therapeutic activity of ECPCM was studied using the mouse strain JR5558, which develops spontaneous choroidal neovascularisation (CNV) lesions driven by local inflammation. RESULTS ECPCM significantly suppressed TNFα-induced expression of E-selectin and VCAM-1. ECPCM did not affect the mRNA stability of the two genes, but suppressed TNFα-induced binding of the p65 subunit of NF-kB transcription factor to E-selectin and VCAM-1 promoters. In vivo, systemic ECPCM treatment significantly reduced the CNV area and the recruitment of activated macrophages to the lesions. Characterization of the molecule responsible for the anti-inflammatory activity in ECPCM indicates that it is unlikely to be a protein and that it is not any of the better characterized EC-derived anti-inflammatory molecules. CONCLUSIONS Medium conditioned by HAEC grown on collagen particles exhibits significant anti-inflammatory activity via inhibition of genes that mediate inflammatory responses in EC.
Collapse
Affiliation(s)
- Laura Paneghetti
- />UCL Institute of Ophthalmology, London, UK
- />Xeptagen S.p.A., Marghera Venice, Italy
| | - Yin-Shan Eric Ng
- />UCL Institute of Ophthalmology, London, UK
- />Schepens Eye Research Institute, 20 Staniford Street, Boston, MA 02114 USA
| |
Collapse
|
10
|
Lee T, Haq NU. New Developments in Our Understanding of Neointimal Hyperplasia. Adv Chronic Kidney Dis 2015; 22:431-7. [PMID: 26524947 DOI: 10.1053/j.ackd.2015.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/26/2015] [Indexed: 12/20/2022]
Abstract
The vascular access remains the lifeline for the hemodialysis patient. The most common etiology of vascular access dysfunction is venous stenosis at the vein-artery anastomosis in arteriovenous fistula and at the vein-graft anastomosis in arteriovenous grafts (AVG). This stenotic lesion is typically characterized on histology as aggressive venous neointimal hyperplasia in both arteriovenous fistula and AVG. In recent years, we have advanced our knowledge and understanding of neointimal hyperplasia in vascular access and begun testing several novel therapies. This article will (1) review recent developments in our understanding of the pathophysiology of neointimal hyperplasia development in AVG and fistula failure, (2) discuss atypical factors leading to neointimal hyperplasia development, (3) highlight key novel therapies that have been evaluated in clinical trials, and (4) discuss future opportunities and challenges to improve our understanding of vascular access dysfunction and translate this knowledge into novel and innovative therapies.
Collapse
|
11
|
Influence of drugs on arteriovenous vascular access dysfunction. J Vasc Access 2015; 16 Suppl 9:S61-5. [PMID: 25751553 DOI: 10.5301/jva.5000365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2015] [Indexed: 01/04/2023] Open
Abstract
Vascular access dysfunction, due to venous stenosis at the vein-artery anastomosis in arteriovenous fistulas and vein-graft anastomosis in synthetic arteriovenous grafts, is a major cause of morbidity and mortality in dialysis patients. The two overarching approaches to prevent and treat vascular access dysfunction are from systemic or local (including endovascular and perivascular) routes. However, there are currently very few effective therapies to treat vascular access dysfunction. This article will review major studies evaluating systemic, endovascular, and perivascular therapies for vascular access dysfunction. Ongoing research to evaluate novel innovations to prevent and/or manage vascular access dysfunction appears promising.
Collapse
|
12
|
Scott R, Panitch A. Macromolecular approaches to prevent thrombosis and intimal hyperplasia following percutaneous coronary intervention. Biomacromolecules 2014; 15:2825-32. [PMID: 24964369 PMCID: PMC4130236 DOI: 10.1021/bm5007757] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/18/2014] [Indexed: 01/29/2023]
Abstract
Cardiovascular disease remains one of the largest contributors to death worldwide. Improvements in cardiovascular technology leading to the current generation of drug-eluting stents, bioresorbable stents, and drug-eluting balloons, coupled with advances in antirestenotic therapeutics developed by pharmaceutical community, have had a profound impact on quality of life and longevity. However, these procedures and devices contribute to both short- and long-term complications. Thus, room for improvement and development of new, alternative strategies exists. Two major approaches have been investigated to improve outcomes following percutaneous coronary intervention including perivascular delivery and luminal paving. For both approaches, polymers play a major role as controlled research vehicles, carriers for cells, and antithrombotic coatings. With improvements in catheter delivery devices and increases in our understanding of the biology of healthy and diseased vessels, the time is ripe for development of novel macromolecular coatings that can protect the vessel lumen following balloon angioplasty and promote healthy vascular healing.
Collapse
Affiliation(s)
- Rebecca
A. Scott
- Weldon
School of Biomedical
Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Alyssa Panitch
- Weldon
School of Biomedical
Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
13
|
Sevestre MA, Larghero J, Castier Y, Nugent HM, Visonneau S, Alsac JM. Pilot safety study of perivascular injection of tissue-engineered allogeneic aortic endothelial cells in patients undergoing minimally invasive peripheral revascularization. J Vasc Surg 2014; 59:1597-606. [PMID: 24613691 DOI: 10.1016/j.jvs.2014.01.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/09/2014] [Accepted: 01/12/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Restenosis is a limitation of endovascular interventions performed in the superficial femoral artery (SFA). Preclinical studies have demonstrated that the perivascular delivery of tissue-engineered allogeneic aortic endothelial cells (PVS-10200) reduced stenosis in porcine models of SFA revascularization. The purpose of this study was to investigate the safety and feasibility of percutaneous PVS-10200 delivery after angioplasty and stenting in the SFA of patients with peripheral artery disease. METHODS In this phase I open-label trial, 21 patients (average lesion length of 10.10 ± 2.36 cm and ≥70% stenosis) were treated with PVS-10200: 11 in a low-dose cohort (cohort A) and 10 in a high-dose cohort (cohort B). The primary objective was to demonstrate the safety (incidence of major adverse events) of PVS-10200 within 4 weeks after surgery. Secondary end points included assessments of resting ankle-brachial index (ABI) in the treated leg, Fontaine class, and time to target lesion revascularization (TLR). RESULTS No patient had a major adverse event within 4 weeks. One patient required a limb amputation at 30 weeks. At 48 weeks, cohort A and cohort B patients maintained a 37% and 62% increase in ABI compared with baseline, respectively; 70% of cohort A and 78% of cohort B improved by ≥1 Fontaine classification stage, and the TLR rate was 39% for cohort A and 20% for cohort B. CONCLUSIONS Percutaneous local delivery of PVS-10200 is a well-tolerated and novel therapeutic approach that may be a suitable treatment for patients after endovascular intervention of the SFA. Larger randomized trials are needed to determine if PVS-10200 can improve ABI and reduce TLR rates.
Collapse
Affiliation(s)
| | - Jerome Larghero
- AP-HP, Saint-Louis Hospital, Cell Therapy Unit and Clinical Investigation in Biotherapies (CBT501), Paris Diderot Sorbonne Paris Cité University, Paris, France
| | - Yves Castier
- AP-HP, Bichat-Claude-Bernard Hospital, Department of Vascular, Thoracic, and Lung Transplantation, Paris
| | | | | | - Jean-Marc Alsac
- AP-HP, Department of Vascular Surgery, Georges Pompidou European Hospital, René Descartes Paris 5 University, Paris, France.
| |
Collapse
|
14
|
High-throughput screening identifies idarubicin as a preferential inhibitor of smooth muscle versus endothelial cell proliferation. PLoS One 2014; 9:e89349. [PMID: 24586708 PMCID: PMC3933427 DOI: 10.1371/journal.pone.0089349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/20/2014] [Indexed: 01/21/2023] Open
Abstract
Intimal hyperplasia is the cause of the recurrent occlusive vascular disease (restenosis). Drugs currently used to treat restenosis effectively inhibit smooth muscle cell (SMC) proliferation, but also inhibit the growth of the protective luminal endothelial cell (EC) lining, leading to thrombosis. To identify compounds that selectively inhibit SMC versus EC proliferation, we have developed a high-throughput screening (HTS) format using human cells and have employed this to screen a multiple compound collection (NIH Clinical Collection). We developed an automated, accurate proliferation assay in 96-well plates using human aortic SMCs and ECs. Using this HTS format we screened a 447-drug NIH Clinical Library. We identified 11 compounds that inhibited SMC proliferation greater than 50%, among which idarubicin exhibited a unique feature of preferentially inhibiting SMC versus EC proliferation. Concentration-response analysis revealed this differential effect most evident over an ∼10 nM-5 µM window. In vivo testing of idarubicin in a rat carotid injury model at 14 days revealed an 80% reduction of intimal hyperplasia and a 45% increase of lumen size with no significant effect on re-endothelialization. Taken together, we have established a HTS assay of human vascular cell proliferation, and identified idarubicin as a selective inhibitor of SMC versus EC proliferation both in vitro and in vivo. Screening of larger and more diverse compound libraries may lead to the discovery of next-generation therapeutics that can inhibit intima hyperplasia without impairing re-endothelialization.
Collapse
|
15
|
Terry CM, Dember LM. Novel therapies for hemodialysis vascular access dysfunction: myth or reality? Clin J Am Soc Nephrol 2013; 8:2202-12. [PMID: 24235283 DOI: 10.2215/cjn.07360713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hemodialysis vascular access dysfunction is a major source of morbidity for patients with ESRD. Development of effective approaches to prevent and treat vascular access failure requires an understanding of the underlying mechanisms, suitable models for preclinical testing, systems for targeted delivery of interventions to maximize efficacy and minimize toxicity, and rigorous clinical trials that use appropriate outcome measures. This article reviews the substantial progress and ongoing challenges in developing novel treatments for arteriovenous vascular access failure and focuses on localized rather than systemic interventions.
Collapse
Affiliation(s)
- Christi M Terry
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, †Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
16
|
Murikipudi S, Methe H, Edelman ER. The effect of substrate modulus on the growth and function of matrix-embedded endothelial cells. Biomaterials 2012; 34:677-84. [PMID: 23102623 DOI: 10.1016/j.biomaterials.2012.09.079] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 09/29/2012] [Indexed: 10/27/2022]
Abstract
Endothelial cells (EC) are potent bioregulatory cells, modulating thrombosis, inflammation and control over mural smooth muscle cells and vascular health. The biochemical roles of EC are retained when cells are embedded within three-dimensional (3D) denatured collagen matrices. Though substrate mechanics have long been known to affect cellular morphology and function and 3D-EC systems are increasingly used as therapeutic modalities little is known about the effect of substrate mechanics on EC in these 3D systems. In this work, we examined the effect of isolated changes in modulus on EC growth and morphology, extracellular matrix gene expression, modulation of smooth muscle cell growth, and immunogenicity. EC growth, but not morphology was dependent on scaffold modulus. Increased scaffold modulus reduced secretion of smooth muscle cell growth inhibiting heparan sulfate proteoglycans (HSPGs), but had no effect on secreted growth factors, resulting in a loss of smooth muscle cell growth inhibition by EC on high modulus scaffolds. Expression of ICAM-1, VCAM-1 and induction of CD4(+) T cell proliferation was reduced by increased scaffold modulus, and correlated with changes in integrin α5 expression. Expression of several common ECM proteins by EC on stiffer substrates dropped, including collagen IV(α1), collagen IV(α5), fibronectin, HSPGs (perlecan and biglycan). In contrast, expression of elastin and TIMPs were increased. This work shows even modest changes in substrate modulus can have a significant impact on EC function in three-dimensional systems. The mechanism of these changes is not clear, but the data presented here within suggests a model wherein EC attempt to neutralize changes in environmental force balance by altering ECM and integrin expression, leading to changes in effects on downstream signaling and function.
Collapse
Affiliation(s)
- Sylaja Murikipudi
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
17
|
Roy-Chaudhury P, El-Khatib M, Campos-Naciff B, Wadehra D, Ramani K, Leesar M, Mistry M, Wang Y, Chan JS, Lee T, Munda R. Back to the Future: How Biology and Technology Could Change the Role of PTFE Grafts in Vascular Access Management. Semin Dial 2012; 25:495-504. [DOI: 10.1111/j.1525-139x.2012.01091.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
18
|
Nugent HM, Ng YS, White D, Groothius A, Kanner G, Edelman ER. Ultrasound-guided percutaneous delivery of tissue-engineered endothelial cells to the adventitia of stented arteries controls the response to vascular injury in a porcine model. J Vasc Surg 2012; 56:1078-88. [PMID: 22796118 DOI: 10.1016/j.jvs.2012.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 02/19/2012] [Accepted: 03/02/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE High restenosis rates are a limitation of peripheral vascular interventions. Previous studies have shown that surgical implantation of a tissue-engineered endothelium onto the adventitia surface of injured vessels regulates vascular repair. In the present study, we developed a particulate formulation of tissue-engineered endothelium and a method to deliver the formulation perivascular to injured blood vessels using a percutaneous, minimally invasive technique. METHODS Stainless steel stents were implanted in 18 balloon-injured femoral arteries of nine domestic swine, followed by ultrasound-guided percutaneous perivascular injection of gelatin particles containing cultured allogeneic porcine aortic endothelial cells (PAE). Controls received injections of empty particles (matrix) or no perivascular injection (sham) after stent deployment. Animals were sacrificed after 90 days. RESULTS Angiographic analysis revealed a significantly greater lumen diameter in the stented segments of arteries treated with PAE/matrix (4.72 ± 0.12 mm) compared with matrix (4.01 ± 0.20 mm) or sham (4.03 ± 0.16 mm) controls (P < .05). Similarly, histologic analysis revealed that PAE/matrix-treated arteries had the greatest lumen area (20.4 ± 0.7 mm(2); P < .05) compared with controls (16.1 ± 0.9 mm(2) and 17.1 ± 1.0 mm(2) for sham and matrix controls, respectively) and the smallest intimal area (3.3 ± 0.4 mm(2); P < .05) compared with controls (6.2 ± 0.5 mm(2) and 4.4 ± 0.5 mm(2) for sham and matrix controls, respectively). Overall, PAE-treated arteries had a 33% to 50% decrease in percent occlusion (P < .05) compared with controls. Histopathological analysis revealed fewer leukocytes present in the intima in the PAE/matrix group compared with control groups, suggesting that the biological effects were in part due to inhibition of the inflammatory phase of the vascular response to injury. CONCLUSIONS Minimally invasive, perivascular delivery of PAE/matrix to stented arteries was performed safely using ultrasound-guided percutaneous injections and significantly decreased stenosis. Application at the time of or subsequent to peripheral interventions may decrease clinical restenosis rates.
Collapse
|
19
|
The role of scaffold microarchitecture in engineering endothelial cell immunomodulation. Biomaterials 2012; 33:7019-27. [PMID: 22796162 DOI: 10.1016/j.biomaterials.2012.06.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/22/2012] [Indexed: 01/13/2023]
Abstract
The implantation of matrix-embedded endothelial cells (MEECs) has been reported to have great therapeutic potential in controlling the vascular response to injury and maintaining patency in arteriovenous anastomoses. While there is an appreciation of their effectiveness in clinical and animal studies, the mechanisms through which they mediate these powerful effects remain relatively unknown. In this work, we examined the hypothesis that the 3-dimensional microarchitecture of the tissue engineering scaffold was a key regulator of endothelial behavior in MEEC constructs. Notably, we found that ECs in porous collagen scaffold had a markedly altered cytoskeletal structure with oriented actin fibers and rearrangement of the focal adhesion proteins in comparison to cells grown on 2D surfaces. We examined the immunomodulatory capabilities of MEECs and discovered that they were able to reduce the recruitment of monocytes to an inflamed endothelial monolayer by 5-fold compared to EC on 2D surfaces. An analysis of secreted factors from the cells revealed an 8-fold lower release of Monocyte Chemotactic Protein-1 (MCP-1) from MEECs. Differences between 3D and 2D cultured cells were abolished in the presence of inhibitors to the focal adhesion associated signaling molecule Src suggesting that adhesion-mediated signaling is essential in controlling the potent immunomodulatory effects of MEEC.
Collapse
|
20
|
De Santis F, Chaves Brait CM, Caravelli G, Pompei S, Di Cintio V. Salvage of infected vascular graft via 'perivascular venous banding' technique coupled with rectus abdominis myocutaneous muscle flap transposition. Vascular 2012; 21:17-22. [PMID: 22375043 DOI: 10.1258/vasc.2011.cr0286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This is the case of a severe Pseudomonas aeruginosa biological vascular graft infection, completely involving the perianastomotic tract of a femoro - femoral crossover bypass and resulting in repeated bleeding from the offended vessel wall. After the failure of a sartorious rotational muscle flap transposition into the infected groin wound, this 'high-grade' vascular graft infection was finally treated successfully by wrapping a great saphenous vein patch reinforcement circumferentially around the damaged biological vascular conduit and filling the infected wound with a rectus abdominis myocutaneous muscle flap transposition. The aim of this report is to illustrate this novel, to our knowledge, 'perivascular venous banding' technique and to evaluate the prospective of future testing of this surgical procedure. Starting from this singular case, we will also review the role of the rotational muscle flaps in the conservative management of major vascular graft infections.
Collapse
Affiliation(s)
- F De Santis
- Department of Vascular Surgery, Sandro Pertini, Rome, Italy.
| | | | | | | | | |
Collapse
|
21
|
Chitalia VC, Murikipudi S, Indolfi L, Rabadi L, Valdez R, Franses JW, Edelman ER. Matrix-embedded endothelial cells are protected from the uremic milieu. Nephrol Dial Transplant 2011; 26:3858-65. [PMID: 21795755 DOI: 10.1093/ndt/gfr337] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Endothelial cells (ECs) embedded in 3D matrices [matrix-embedded endothelial cells (MEECs)] of denatured collagen implanted around vascular access anastomoses preserve luminal patency. MEEC implant efficacy depends on embedded EC health. As the uremic milieu inhibits proliferation and induces apoptosis of ECs, we examined whether uremia might impact MEECs. METHODS ECs grown on 2D gelatin-coated polystyrene tissue culture plates (gTCPS) or in MEEC were treated with sera pooled from 20 healthy control or uremic patients with end-stage renal disease. EC viability was examined using 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide assay, cell counting and Trypan blue exclusion. Media conditioned (CM) with 2 and 3D-supported ECs were examined for its potential to inhibit vascular smooth muscle cell (vSMC) proliferation using (3)[H] thymidine incorporation and cyclin D1 expression. ECs grown on gTCPS were treated with uremic serum filtered through matrices to examine if matrices retain uremic toxins or whether EC effects were cell mediated. RESULTS Uremic serum significantly reduced viability and number of live, and increased dead ECs when grown on gTCPS, but not in MEECs. EC survival correlated with vSMC inhibition. While CM from ECs grown in gTCPS with uremic serum inhibited vSMC proliferation no better than uremic serum alone (22 versus 27%), MEEC CM inhibited vSMC proliferation by 47% (P = 0.0004). Cyclin D1 expression tracked with indices of vSMC proliferation. There was no significant difference in EC viability between EC treated with matrix-filtered or unfiltered uremic serum. CONCLUSION The viability, number and efficacy of MEECs were preserved in uremic serum compared to those of ECs on gTCPS. MEECs are protected from uremic toxicity, not from retention of uremic toxins by matrices, but likely from intrinsic changes in EC sensitivity to uremia. MEECs implanted at vascular access sites should inhibit neointimal hyperplasia in uremia. This study underscores the robustness of matrix embedding as a cell protectant, especially in hostile environments like uremia.
Collapse
Affiliation(s)
- Vipul C Chitalia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Harnek J, Zoucas E, de Sá VP, Ekblad E, Arner A, Stenram U. Intimal hyperplasia in balloon dilated coronary arteries is reduced by local delivery of the NO donor, SIN-1 via a cGMP-dependent pathway. BMC Cardiovasc Disord 2011; 11:30. [PMID: 21663688 PMCID: PMC3123303 DOI: 10.1186/1471-2261-11-30] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 06/11/2011] [Indexed: 02/05/2023] Open
Abstract
Background To elucidate the mechanism by which local delivery of 3-morpholino-sydnonimine (SIN-1) affects intimal hyperplasia after percutaneous transluminal coronary angioplasty (PTCA). Methods Porcine coronary arteries were treated with PTCA and immediately afterwards locally treated for 5 minutes, with a selective cytosolic guanylate cyclase inhibitor, 1 H-(1,2,4)oxadiazole(4,3-alpha)quinoxaline-1-one (ODQ) + SIN-1 or only SIN-1 using a drug delivery-balloon. Arteries were angiographically depicted, morphologically evaluated and analyzed after one and eight weeks for actin, myosin and intermediate filaments (IF) and nitric oxide synthase (NOS) contents. Results Luminal diameter after PCI in arteries treated with SIN-1 alone and corrected for age-growth was significantly larger as compared to ODQ + SIN-1 or to controls (p < 0.01). IF/actin ratio after one week in SIN-1 treated segments was not different compared to untreated segments, but was significantly reduced compared to ODQ + SIN-1 treated vessels (p < 0.05). Expression of endothelial NADPH diaphorase activity was significantly lower in untreated segments and in SIN-1 treated segments compared to controls and SIN-1 + ODQ treated arteries (p < 0.01). Restenosis index (p < 0.01) and intimal hyperplasia (p < 0.01) were significantly reduced while the residual lumen was increased (p < 0.01) in SIN-1 segments compared to controls and ODQ + SIN-1 treated vessels. Conclusions After PTCA local delivery of high concentrations of the NO donor SIN-1 for 5 minutes inhibited injury induced neointimal hyperplasia. This favorable effect was abolished by inhibition of guanylyl cyclase indicating mediation of a cyclic guanosine 3',5'-monophosphate (cGMP)-dependent pathway. The momentary events at the time of injury play crucial role in the ensuring development of intimal hyperplasia.
Collapse
Affiliation(s)
- Jan Harnek
- Department of Coronary Heart Disease, Skane University Hospital, Institute of Clinical Sciences, Lunds University, Getingev 4, SE-22185 Lund, Sweden.
| | | | | | | | | | | |
Collapse
|
23
|
T-helper 2 cells are essential for modulation of vascular repair by allogeneic endothelial cells. J Heart Lung Transplant 2009; 29:479-86. [PMID: 20036161 DOI: 10.1016/j.healun.2009.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 11/08/2009] [Accepted: 11/09/2009] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Endothelial cells (ECs) embedded within 3-dimensional matrices (MEEC) control lumenal inflammation and intimal hyperplasia when placed in the vascular adventitia. Matrix embedding alters endothelial immunogenicity in vitro. T-helper (Th) cell-driven host immunity is an impediment of allogeneic grafts. We aimed to identify if modulation of Th balance would affect immune compatibility and endothelial regulation of vascular repair in vivo. METHODS Pigs (n = 4/group) underwent carotid artery balloon injury and were left untreated (Group 1) or received perivascular porcine MEEC implants (Group 2), 12 days of cyclosporine A (CsA; Group 3), or MEEC and CsA (Group 4). Host immune reactivity was analyzed after 28 and 90 days. RESULTS MEEC treatment induced formation of EC-specific immunoglobulin (Ig) G(1) antibodies (41 +/- 6 mean fluorescence intensity [MFI]) and differentiation of host splenocytes into Th2, but not Th1, cytokine-producing cells (interleukin [IL]-4, 242 +/- 102; IL-10, 273 +/- 114 number of spots). Concomitant CsA therapy reduced IgG(1) antibody frequency (25 +/- 2 MFI; p < 0.02) and Th2-cytokine producing splenocytes upon MEEC treatment (IL-4, 157 +/- 19; IL-10, 124 +/- 26 number of spots; p < 0.05). MEECs inhibited luminal occlusion 28 and 90 days after balloon injury (12 +/- 7%) vs untreated controls (68 +/- 14%; p < 0.001) but to a lesser extent with concomitant CsA treatment (34 +/- 13%; p < 0.02 vs Group 2). CONCLUSIONS MEECs do not induce a significant Th1-driven immune response but do enhance differentiation of splenocytes into cells producing Th2 cytokine. Reduction in this Th2 response reduces the vasoregulatory effects of allogeneic ECs after injury.
Collapse
|
24
|
Conte MS, Nugent HM, Gaccione P, Guleria I, Roy-Chaudhury P, Lawson JH. Multicenter phase I/II trial of the safety of allogeneic endothelial cell implants after the creation of arteriovenous access for hemodialysis use: The V-HEALTH study. J Vasc Surg 2009; 50:1359-68.e1. [DOI: 10.1016/j.jvs.2009.07.108] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2009] [Revised: 07/21/2009] [Accepted: 07/25/2009] [Indexed: 11/15/2022]
|
25
|
Delivery site of perivascular endothelial cell matrices determines control of stenosis in a porcine femoral stent model. J Vasc Interv Radiol 2009; 20:1617-24. [PMID: 19854069 DOI: 10.1016/j.jvir.2009.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 06/30/2009] [Accepted: 08/26/2009] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Endothelial cells, grown within gelatin matrices and implanted onto the adventitia of injured vessels, inhibit stenosis in experimental models. To determine if this technology could be adapted for minimally invasive procedures, the authors compared the effects of cells in an implantable sponge to that of an injectable formulation and investigated the importance of delivery site in a stent model. MATERIALS AND METHODS Stents were implanted in the femoral arteries of 30 pigs. This was followed by perivascular implantation of sponges or injection of particles containing allogeneic endothelial cells. Controls received acellular matrices or nothing. The effects of delivery site were assessed by injecting cellular matrices into or adjacent to the perivascular tissue or into the neighboring muscle. Animals were sacrificed after 28 days. Pre-sacrifice angiograms and tissue sections were evaluated for stenosis. RESULTS Arteries treated with cellular matrices had a 55%-63% decrease in angiographic stenosis (P < .05) and a 38%-43% reduction in histologic stenoses (P < .05) compared to controls. Intimal area was greatest when cellular matrices were delivered into the muscle (6.35 mm(2) +/- 0.95) rather than into or adjacent to the perivascular tissue (4.05 mm(2) +/- 0.56 and 4.73 mm(2) +/- 0.53, respectively; P < .05). CONCLUSIONS Perivascular endothelial cell matrices reduced stenosis after stent-induced injury. The effects were not dependent on the formulation but appeared to be dependent on delivery site. Minimally invasive injections of endothelial cell matrices to the adventitia of arteries following peripheral interventions may decrease restenosis rates.
Collapse
|
26
|
Kenagy RD, Fukai N, Min SK, Jalikis F, Kohler TR, Clowes AW. Proliferative capacity of vein graft smooth muscle cells and fibroblasts in vitro correlates with graft stenosis. J Vasc Surg 2009; 49:1282-8. [PMID: 19307078 DOI: 10.1016/j.jvs.2008.12.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 12/04/2008] [Accepted: 12/07/2008] [Indexed: 11/18/2022]
Abstract
OBJECTIVE About a quarter of peripheral vein grafts fail due in part to intimal hyperplasia. The proliferative capacity and response to growth inhibitors of medial smooth muscle cells and adventitial fibroblasts in vitro were studied to test the hypothesis that intrinsic differences in cells of vein grafts are associated with graft failure. METHODS Cells were grown from explants of the medial and adventitial layers of samples of vein grafts obtained at the time of implantation. Vein graft patency and function were monitored over the first 12 months using ankle pressures and Duplex ultrasound to determine vein graft status. Cells were obtained from veins from 11 patients whose grafts remained patent (non-stenotic) and from seven patients whose grafts developed stenosis. Smooth muscle cells (SMCs) derived from media and fibroblasts derived from adventitia were growth arrested in serum-free medium and then stimulated with 1 muM sphingosine-1-phosphate (S1P), 10 nM thrombin, 10 ng/ml epidermal growth factor (EGF), 10 ng/ml platelet-derived growth factor-BB (PDGF-BB), PDGF-BB plus S1P, or PDGF-BB plus thrombin for determination of incorporation of [(3)H]-thymidine into DNA. Cells receiving PDGF-BB or thrombin were also treated with or without 100 microg/ml heparin, which is a growth inhibitor. Cells receiving thrombin were also treated with or without 150 nM AG1478, an EGF receptor kinase inhibitor. RESULTS SMCs and fibroblasts from veins of patients that developed stenosis responded more to the growth factors, such as PDGF-BB alone or in combination with thrombin or S1P, than cells from veins of patients that remained patent (P = .012). In addition, while PDGF-BB-mediated proliferation of fibroblasts from grafts that remained patent was inhibited by heparin (P < .03), PDGF-BB-mediated proliferation of fibroblasts from veins that developed stenosis was not (P > .5). CONCLUSION Inherent differences in the proliferative response of vein graft cells to PDGF-BB and heparin may explain, in part, the variability among patients regarding long term patency of vein grafts.
Collapse
MESH Headings
- Aged
- Ankle/blood supply
- Becaplermin
- Blood Pressure
- Cell Proliferation/drug effects
- Cells, Cultured
- Constriction, Pathologic
- DNA Replication
- Epidermal Growth Factor/metabolism
- Female
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Graft Occlusion, Vascular/etiology
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/physiopathology
- Heparin/pharmacology
- Humans
- Hyperplasia
- Lower Extremity/blood supply
- Lysophospholipids/metabolism
- Male
- Middle Aged
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Peripheral Vascular Diseases/pathology
- Peripheral Vascular Diseases/physiopathology
- Peripheral Vascular Diseases/surgery
- Platelet-Derived Growth Factor/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-sis
- Quinazolines
- Saphenous Vein/drug effects
- Saphenous Vein/pathology
- Saphenous Vein/physiopathology
- Saphenous Vein/transplantation
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Thrombin/metabolism
- Time Factors
- Tyrphostins/pharmacology
- Ultrasonography, Doppler, Duplex
- Vascular Patency
Collapse
Affiliation(s)
- Richard D Kenagy
- Department of Surgery, University of Washington Medical School, Seattle, Wash. 98195-6410, USA
| | | | | | | | | | | |
Collapse
|
27
|
Gulati R, Simari RD. Defining the potential for cell therapy for vascular disease using animal models. Dis Model Mech 2009; 2:130-7. [PMID: 19259386 PMCID: PMC2650189 DOI: 10.1242/dmm.000562] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cell-based therapeutics are currently being developed for a wide array of unmet medical needs. As obstructive vascular disease is the major cause of mortality in the world, cell-based strategies aimed at developing novel therapies or improving current therapies are currently under study. These studies are based on the evolving understanding of the biology of vascular progenitor cells, which has in turn led to the availability of well-defined sources of vascular cells for delivery. Crucial to the development of these approaches is the preclinical testing of cell delivery in animal models. This review highlights the crucial steps involved in the selection of cell sources and generation, delivery approaches, animal models to be used, and endpoints to be studied, in the context of cell delivery for vascular disease. Furthermore, the development of cell delivery to induce angiogenesis in ischemic limbs and to improve the response to large vessel injury will be discussed.
Collapse
Affiliation(s)
- Rajiv Gulati
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert D. Simari
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
28
|
Hess S, Methe H, Kim JO, Edelman ER. NF-kappaB activity in endothelial cells is modulated by cell substratum interactions and influences chemokine-mediated adhesion of natural killer cells. Cell Transplant 2009; 18:261-73. [PMID: 19558775 PMCID: PMC2857529 DOI: 10.3727/096368909788534979] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Because changes in subendothelial matrix composition are associated with alterations of the endothelial immune phenotype, we sought to understand if cytokine-induced NF-kappaB activity and downstream effects depend on substrate adherence of endothelial cells (EC). We compared the upstream phosphorylation cascade, activation of NF-kappaB, and expression/secretion of downstream effects of EC grown on tissue culture polystyrene plates (TCPS) with EC embedded within collagen-based matrices (MEEC). Adhesion of natural killer (NK) cells was quantified in vitro and in vivo. NF-kappaB subunit p65 nuclear levels were significantly lower and p50 significantly higher in cytokine-stimulated MEEC than in EC-TCPS. Despite similar surface expression of TNF-alpha receptors, MEEC had significantly decreased secretion and expression of IL-6, IL-8, MCP-1, VCAM-1, and ICAM-1. Attenuated fractalkine expression and secretion in MEEC (two to threefold lower than in EC-TCPS; p < 0.0002) correlated with 3.7-fold lower NK cell adhesion to EC (6,335 +/- 420 vs. 1,735 +/- 135 cpm; p < 0.0002). Furthermore, NK cell infiltration into sites of EC implantation in vivo was significantly reduced when EC were embedded within matrix. Matrix embedding enables control of EC substratum interaction. This in turn regulates chemokine and surface molecule expression and secretion, in particular of those compounds within NF-kappaB pathways, chemoattraction of NK cells, local inflammation, and tissue repair.
Collapse
Affiliation(s)
- Shmuel Hess
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | |
Collapse
|
29
|
Tissue-engineered endothelial and epithelial implants differentially and synergistically regulate airway repair. Proc Natl Acad Sci U S A 2008; 105:7046-51. [PMID: 18458330 DOI: 10.1073/pnas.0802463105] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The trilaminate vascular architecture provides biochemical regulation and mechanical integrity. Yet regulatory control can be regained after injury without recapitulating tertiary structure. Tissue-engineered (TE) endothelium controls repair even when placed in the perivascular space of injured vessels. It remains unclear from vascular repair studies whether endothelial implants recapitulate the vascular epithelial lining or expose injured tissues to endothelial cells (ECs) with unique healing potential because ECs line the vascular epithelium and the vasa vasorum. We examined this issue in a nonvascular tubular system, asking whether airway repair is controlled by bronchial epithelial cells (EPs) or by ECs of the perfusing bronchial vasculature. Localized bronchial denuding injury damaged epithelium, narrowed bronchial lumen, and led to mesenchymal cell hyperplasia, hypervascularity, and inflammatory cell infiltration. Peribronchial TE constructs embedded with EPs or ECs limited airway injury, although optimum repair was obtained when both cells were present in TE matrices. EC and EP expression of PGE(2), TGFbeta1, TGFbeta2, GM-CSF, IL-8, MCP-1, and soluble VCAM-1 and ICAM-1 was altered by matrix embedding, but expression was altered most significantly when both cells were present simultaneously. EPs may provide for functional control of organ injury and fibrous response, and ECs may provide for preservation of tissue perfusion and the epithelium in particular. Together the two cells optimize functional restoration and healing, suggesting that multiple cells of a tissue contribute to the differentiated biochemical function and repair of a tissue, but need not assume a fixed, ordered architectural relationship, as in intact tissues, to achieve these effects.
Collapse
|
30
|
Methe H, Hess S, Edelman ER. The effect of three-dimensional matrix-embedding of endothelial cells on the humoral and cellular immune response. Semin Immunol 2008; 20:117-22. [PMID: 18243732 DOI: 10.1016/j.smim.2007.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 12/10/2007] [Accepted: 12/10/2007] [Indexed: 10/22/2022]
Abstract
The endothelium is a unique immunologic target. The first host-donor reaction in any cell, tissue or organ transplant occurs at the blood-tissue interface, the endothelium. When endothelial cells are themselves the primary component of the implant a second set of immunologic reactions arises. Injections of free endothelial cell implants elicit a profound major histocompatibility complex (MHC) II dominated immune response with significant sensitivity, cascade enhancement and immune memory. Endothelial cells embedded within three-dimensional matrices retain all the biosecretory capacity of quiescent endothelial cells. Perivascular implants of such cells are the most potent inhibitor of intimal hyperplasia and thrombosis following controlled vascular injury, but without any immune reactivity. Allo- and even xenogeneic endothelial cells evoke no significant humoral or cellular immune response in immunocompetent hosts when embedded within matrices. Moreover, endothelial implants are immunomodulatory, reducing the extent of the memory response to previous free cell implants. Attenuated immunogenicity results in muted activation of adaptive and innate immune cells. These findings point toward a pivotal role of matrix-cell-interconnectivity for the cellular immune phenotype and might therefore assist in the design of extracellular matrix components for successful tissue engineering.
Collapse
Affiliation(s)
- Heiko Methe
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | |
Collapse
|
31
|
Nugent HM, Sjin RTT, White D, Milton LG, Manson RJ, Lawson JH, Edelman ER. Adventitial endothelial implants reduce matrix metalloproteinase-2 expression and increase luminal diameter in porcine arteriovenous grafts. J Vasc Surg 2007; 46:548-556. [PMID: 17826244 PMCID: PMC2702136 DOI: 10.1016/j.jvs.2007.04.074] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 04/25/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Vascular access dysfunction is a major problem in hemodialysis patients. Only 50% of arteriovenous grafts (AVGs) will remain patent 1 year after surgery. AVGs frequently develop stenoses and occlusions at the venous anastomoses in the venous outflow tract. Lumen diameter is not only determined by intimal thickening but is also influenced by remodeling of the vessel wall. Vascular remodeling requires degradation and reorganization of the extracellular matrix by the degradation enzymes, matrix metalloproteinases (MMPs). In this study, we aimed to provide further insight into the mechanism of endothelial regulation of vascular remodeling and luminal narrowing in AVGs. METHODS End-to-side carotid artery-jugular vein polytetrafluoroethylene grafts were created in 20 domestic swine. The anastomoses and outflow vein were treated with Gelfoam matrices (Pfizer, New York, NY) containing allogeneic porcine aortic endothelial (PAE, n = 10) cells or control matrices without cells (n = 10), and the biologic responses to PAE implants were investigated 3 and 28 days postoperatively. Angiograms before euthanasia were compared with baseline angiograms. Tissue sections were stained with hematoxylin and eosin, Verhoeff elastin, and antibodies specific to MMP-9 and MMP-2 and underwent histopathologic, morphometric and immunohistochemical analysis. RESULTS Veins treated with PAE cell implants had a 2.8-fold increase in venous lumen diameter compared with baseline (P < .05), a 2.3-fold increase in lumen diameter compared with control, and an 81% decrease in stenosis (P < .05) compared with control at 28 days. The increase in lumen diameter by angiographic analysis correlated with morphometric analysis of tissue sections. PAE implants increased the venous lumen area 2.3-fold (P < .05), decreased venous luminal occlusion 66%, and increased positive venous remodeling 1.9-fold (P < .05) compared with control at 28 days. PAE cell implants reduced MMP-2 expression and neovascularization at 3 and 28 days and adventitial fibrosis at 28 days, suggesting a role of the implants in controlling the affects of medial and adventitial cells in the response to vascular injury. CONCLUSIONS These results demonstrate that the adventitial application of endothelial implants significantly reduced MMP-2 expression within the venous wall, and increased venous lumen diameter and positive remodeling in a porcine arteriovenous graft model. Adventitial endothelial implants may be useful in decreasing luminal narrowing in a clinical setting.
Collapse
Affiliation(s)
- Helen M Nugent
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
BACKGROUND While tissue engineering offers promise for organ and tissue transplantation, it can also be used to examine transplant and immune biology. Endothelial cells engrafted within 3-dimensional matrices create stable units that produce all of the factors of a functional quiescent endothelium. Perivascular implantation of tissue engineered endothelial cell constructs provides long-term control of vascular repair after injury. This control is established without restoration of the natural luminal:mural endothelium, and most intriguingly, without engendering host allo- and xenogeneic immune responses. We examined how endothelial immunogenicity is controlled by interaction with 3-dimensional matrices. MATERIALS AND METHODS Human aortic endothelial cells (HAE) were either grown to confluence on polystyrene tissue culture plates or within 3-dimensional collagen-based matrices. Major histocompatibility complex (MHC) class II, integrin, interferon (IFN)-gamma receptor expression, and signaling were analyzed via confocal microscopy, flow cytometry, reverse transcription polymerase chain reaction (RT-PCR), and microarray. Splenocyte proliferation was assayed by thymidine incorporation. RESULTS Despite similar expression levels of IFN-gamma receptors, matrix-embedded HAE elicited far less STAT-1 phosphorylation upon IFN-gamma stimulation, and expressed 2-fold less MHC II than HAE grown to confluence on culture plates (P < .001). This effect correlated with reduced expression of integrin alpha(v) and beta(3) (P < .002), and muted proliferation of porcine splenocytes (P < .001). CONCLUSIONS Matrix architecture is critical for modulation of endothelial immunogenicity. Embedding HAE within a physiologic 3-dimensional environment affects activity of intracellular signaling pathways, MHC II expression, and subsequent activation of immune cells. These findings might offer novel insights into our understanding of endothelial-mediated diseases and might enhance our ability to leverage the potential for cell-based therapies.
Collapse
Affiliation(s)
- H Methe
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | |
Collapse
|
33
|
Methe H, Groothuis A, Sayegh MH, Edelman ER. Matrix adherence of endothelial cells attenuates immune reactivity: induction of hyporesponsiveness in allo‐ and xenogeneic models. FASEB J 2007; 21:1515-26. [PMID: 17264166 DOI: 10.1096/fj.06-7051com] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial integrity regulates vascular tone, luminal patency, and the immune reactivity to tissue grafts. Endothelial dysfunction is the first marker and site of disease initiation and severity. It has long been known that endothelial biochemical function is density dependent, and we have recently shown that endothelial immunobiology is anchorage dependent. Matrix-embedded endothelial cells (EC) establish a controlled anchorage state and are not only immune protected but also induce a system immune protective state. We now define this aspect of vascular and immune biology in detail. The in vitro immune response of allogeneic splenocytes (proliferation, lytic activity, and cytokine expression) on exposure to aortic EC was significantly reduced if EC were embedded within three-dimensional collagen matrices (3D-EC; P<0.005) to an even greater extent than EC that had reached confluence as monolayers on tissue culture plates (EC-TCPS). Splenocyte reactivity was enhanced with repeated exposure to EC-TCPS but minimally if preexposed to 3D-EC (P<0.002). 3D-EC induced significantly greater differentiation of splenocytes into CD4+ CD25+ Foxp3+ regulatory T cells than EC-TCPS (P<0.02). The reduced response to 3D-EC and potential protective effect to subsequent exposure were confirmed in vivo. Repeated exposure of immune-competent mice to injections of xenogeneic EC-TCPS induced vigorous host immunity. In contrast, prior implantation of 3D-EC induced hyporesponsiveness toward subsequent injection of EC-TCPS with reduced humoral response, decreased lytic activity, and lower frequency of effector splenocytes (P<0.001). EC interaction with its matrix determines phenotype, viability, and biosecretory potential. We now show that this microenvironmental interaction also influences endothelial-mediated activation of allo- and xenogeneic immune cells. 3D matrix-embedding limits the ability of EC to initiate adaptive immunity, and initial exposure to 3D-EC confers hyporesponsiveness to subsequent exposure to immunogeneic EC. These effects transcended the traditional control that confluence imposes on EC and reflects perhaps even higher order control. Our findings might offer novel insights to endothelial-mediated diseases and potential cell-based therapies.
Collapse
Affiliation(s)
- Heiko Methe
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Ave., Bldg 56-322, Cambridge, MA 02139, USA.
| | | | | | | |
Collapse
|
34
|
|
35
|
Mazighi M, Tchétché D, Gouëffic Y, San Juan A, Louedec L, Hénin D, Michel JB, Jacob MP, Feldman LJ. Percutaneous transplantation of genetically-modified autologous fibroblasts in the rabbit femoral artery: a novel approach for cardiovascular gene therapy. J Vasc Surg 2006; 44:1067-1075. [PMID: 17098544 DOI: 10.1016/j.jvs.2006.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Accepted: 07/06/2006] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Arterial cell and gene therapies are promising strategies for the treatment of cardiovascular diseases; however, the optimal cell type and delivery technique for such treatment remain to be determined. The aim of the present study was to design a new approach for arterial cell and gene therapy in which genetically modified autologous skin fibroblasts are percutaneously delivered in stented rabbit femoral arteries in vivo. METHODS Autologous skin fibroblasts underwent in vitro transfection with the cationic lipid FuGene and plasmids expressing the human form of the tissue inhibitor of metalloproteinase (hTIMP-1) or nls-LacZ reporter genes. RESULT Transfection efficiency was about 50% and there were high levels of hTIMP-1 secretion up to 14 days after gene transfer. We demonstrated the feasibility of in vivo percutaneous transplantation of fluorescent fibroblasts in the rabbit femoral artery. Results were confirmed by scanning electron microscopy. In vivo local delivery of hTIMP-1-expressing fibroblasts in stented femoral arteries also resulted in high-levels of hTIMP-1 secretion ex vivo for 7 days. Fibroblast transplantation resulted in a modest increase in intimal hyperplasia at the target site, which was reversed with hTIMP-1-transfected fibroblasts. CONCLUSION Percutaneous transplantation of genetically modified autologous fibroblasts could be used as a cellular platform for locoregional secretion of therapeutic proteins to treat either specific arterial diseases or the diseased organ (eg, the heart) supplied by the target artery. CLINICAL RELEVANCE Cell and gene therapies are potential new treatments for cardiovascular diseases. We demonstrated that autologous fibroblasts could be easily harvested from a skin biopsy specimen, genetically modified in vitro with nonviral vectors, and percutaneously seeded in vivo in rabbit femoral arteries, leading to locoregional secretion of abundant amounts of recombinant proteins. This new approach has important advantages over alternative approaches that use endothelial cells, viral vectors, and intraoperative cell delivery. Clinical applications may include local treatment of atherosclerotic plaques or aneurysms and also treatment of the diseased organs supplied by the target artery (eg, ischemic or failing heart).
Collapse
|
36
|
Pislaru SV, Harbuzariu A, Gulati R, Witt T, Sandhu NP, Simari RD, Sandhu GS. Magnetically targeted endothelial cell localization in stented vessels. J Am Coll Cardiol 2006; 48:1839-45. [PMID: 17084259 DOI: 10.1016/j.jacc.2006.06.069] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 06/26/2006] [Accepted: 06/26/2006] [Indexed: 01/22/2023]
Abstract
OBJECTIVES A novel method to magnetically localize endothelial cells at the site of a stented vessel wall was developed. The application of this strategy in a large animal model is described. BACKGROUND Local delivery of blood-derived endothelial cells has been shown to facilitate vascular healing in animal models. Therapeutic utilization has been limited by an inability to retain cells in the presence of blood flow. We hypothesized that a magnetized stent would facilitate local retention of superparamagnetically labeled cells. METHODS Cultured porcine endothelial cells were labeled with endocytosed superparamagnetic iron oxide microspheres. A 500:1 microsphere-to-cell ratio was selected for in vivo experiments based on bromo-deoxyuridine incorporation and terminal deoxynucleotidyl transferase mediated dUTP nick end labeling assays. Stents were magnetized and implanted in porcine coronary and femoral arteries using standard interventional equipment. Labeled endothelial cells were delivered locally during transient occlusion of blood flow. RESULTS The delivered cells were found attached to the stent struts and were also distributed within the adjacent denuded vessel wall at 24 h. CONCLUSIONS Magnetic forces can be used to rapidly place endothelial cells at the site of a magnetized intravascular stent. The delivered cells are retained in the presence of blood flow and also spread to the adjacent injured vessel wall. Potential applications include delivering a cell-based therapeutic effect to the local vessel wall as well as downstream tissue.
Collapse
Affiliation(s)
- Sorin V Pislaru
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Roy-Chaudhury P, Sukhatme VP, Cheung AK. Hemodialysis vascular access dysfunction: a cellular and molecular viewpoint. J Am Soc Nephrol 2006; 17:1112-27. [PMID: 16565259 DOI: 10.1681/asn.2005050615] [Citation(s) in RCA: 416] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hemodialysis vascular access dysfunction is a major cause of morbidity and hospitalization in the hemodialysis population. The major cause of hemodialysis vascular access dysfunction is venous stenosis as a result of neointimal hyperplasia. Despite the magnitude of the clinical problem, however, there has been a paucity of novel therapeutic interventions in this field. This is in marked contrast to a recent plethora of targeted interventions for the treatment of arterial neointimal hyperplasia after coronary angioplasty. The reasons for this are two-fold. First there has been a relative lack of cellular and molecular research that focuses on venous neointimal hyperplasia in the specific setting of hemodialysis vascular access. Second, there have been inadequate efforts by the nephrology community to translate the recent advances in molecular and interventional cardiology into therapies for hemodialysis vascular access. This review therefore (1) briefly examines the different forms of hemodialysis vascular access that are available, (2) describes the pathology and pathogenesis of hemodialysis vascular access dysfunction in both polytetrafluoroethylene grafts and native arteriovenous fistulae, (3) reviews recent concepts about the pathogenesis of vascular stenosis that could potentially be applied in the setting of hemodialysis vascular access dysfunction, (4) summarizes novel experimental and clinical therapies that could potentially be used in the setting of hemodialysis vascular access dysfunction, and, finally, (5) offers some broad guidelines for future innovative translational and clinical research in this area that hopefully will reduce the huge clinical morbidity and economic costs that are associated with this condition.
Collapse
Affiliation(s)
- Prabir Roy-Chaudhury
- Division of Nephrology, MSB G-251, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0585.
| | | | | |
Collapse
|
38
|
Yow KH, Ingram J, Korossis SA, Ingham E, Homer-Vanniasinkam S. Tissue engineering of vascular conduits. Br J Surg 2006; 93:652-61. [PMID: 16703652 DOI: 10.1002/bjs.5343] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Autologous conduits are not available in up to 40 per cent of patients with arteriopathy who require coronary or lower limb revascularization, and access sites for renal dialysis may eventually become exhausted. Synthetic prostheses achieve a poor patency rate in small-calibre anastomoses. This review examines how vascular tissue engineering may be used to address these issues. METHODS A Medline search was performed, using the keywords "vascular tissue engineering", "small diameter vascular conduit", "vascular cell biology", "biomechanics", "cell seeding" and "graft endothelialization". Key references were hand-searched for relevant papers. RESULTS AND CONCLUSION In vitro and in vivo approaches are currently being used for guided cell repopulation of both biological and synthetic scaffolds. The major clinical problem has been extended culture time (approximately 6 weeks), which precludes their use in the acute setting. However, recent advances have led not only to improved patency rates for prostheses, but also to a potential reduction in culture time. In addition, increased mobilization of endothelial progenitor cells in the presence of ischaemic tissue may increase the autologous cell yield for scaffold reseeding with further reduction in culture time.
Collapse
Affiliation(s)
- K-H Yow
- Institute of Medical and Biological Engineering, University of Leeds, Leeds General Infirmary, Leeds, UK.
| | | | | | | | | |
Collapse
|
39
|
Brewster L, Brey E, Greisler H. Cardiovascular gene delivery: The good road is awaiting. Adv Drug Deliv Rev 2006; 58:604-29. [PMID: 16769148 PMCID: PMC3337725 DOI: 10.1016/j.addr.2006.03.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 03/24/2006] [Indexed: 01/13/2023]
Abstract
Atherosclerotic cardiovascular disease is a leading cause of death worldwide. Despite recent improvements in medical, operative, and endovascular treatments, the number of interventions performed annually continues to increase. Unfortunately, the durability of these interventions is limited acutely by thrombotic complications and later by myointimal hyperplasia followed by progression of atherosclerotic disease over time. Despite improving medical management of patients with atherosclerotic disease, these complications appear to be persisting. Cardiovascular gene therapy has the potential to make significant clinical inroads to limit these complications. This article will review the technical aspects of cardiovascular gene therapy; its application for promoting a functional endothelium, smooth muscle cell growth inhibition, therapeutic angiogenesis, tissue engineered vascular conduits, and discuss the current status of various applicable clinical trials.
Collapse
Affiliation(s)
- L.P. Brewster
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - E.M. Brey
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
- Research and Surgical Services, Edward J. Hines Jr. V.A. Hospital, Hines, IL, 60141, USA
| | - H.P. Greisler
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Medical Center, Maywood, IL, 60153, USA
- Research and Surgical Services, Edward J. Hines Jr. V.A. Hospital, Hines, IL, 60141, USA
- Corresponding author. Loyola University Medical Center, Department of Surgery, 2160 South First Avenue, Maywood, IL, 60153, USA. Tel.: +1 708 216 8541; fax: +1 708 216 6300. (H.P. Greisler)
| |
Collapse
|
40
|
Gulati R, Lerman A, Simari RD. Therapeutic uses of autologous endothelial cells for vascular disease. Clin Sci (Lond) 2005; 109:27-37. [PMID: 15966869 DOI: 10.1042/cs20050002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Endothelial cells play important structural and functional roles in vascular homoeostasis. Perturbations in endothelial cell number and function are directly involved with the initiation and progression of multiple cardiovascular diseases, including atherosclerosis, hypertension and congestive heart failure. Attempts to modify these disorders have included pharmacological strategies to improve vascular and thus endothelial function. A goal of biological approaches to these disorders is the delivery of endothelial cells that might act to provide beneficial endothelial-derived factors. However, this approach has generally been limited by the lack of readily available autologous endothelial cells for delivery. The isolation of circulation-derived endothelial progenitor cells allows for direct access to autologous endothelial cells for preclinical and clinical studies. Preclinical studies using autologous endothelial cells have demonstrated beneficial effects when delivered in animal models of vascular injury and grafting. These effects are related to the endothelial nature of the cells and may be paracrine in nature. Ongoing studies are aimed at defining the nature of these effects and optimizing delivery strategies cognizant of these mechanisms.
Collapse
Affiliation(s)
- Rajiv Gulati
- Department of Cardiovascular Medicine, University of Birmingham, Birmingham, UK
| | | | | |
Collapse
|
41
|
Solanes N, Rigol M, Ramírez J, Segalés J, Roqué M, Marimon JM, Pérez-Villa F, Roig E, Sanz G, Heras M. Histological Basis of the Porcine Femoral Artery for Vascular Research. Anat Histol Embryol 2005; 34:105-11. [PMID: 15771672 DOI: 10.1111/j.1439-0264.2004.00580.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The similarities between the porcine and human cardiovascular systems make the pig a useful animal model for the study of vascular biology. However, a standardized method is needed to describe the normal histological properties of porcine arteries in order to evaluate pathologic lesions in future studies. Descriptive and morphometric analyses were done on 16 porcine femoral arteries. For these purposes, three histological stains (haematoxylin eosin, Masson's trichrome, and orcein), four immunohistochemical methods (using antibodies anti-alpha-actin, anti-CD3, anti-L1 and anti-lysozyme), and a glycohistochemical method (using Dolichos biflorus lectin) were performed. The porcine femoral arteries evaluated had a mean total area of 6.25 +/- 1.99 mm(2) and a diameter of 2.79 +/- 0.41 mm. The majority of the total area was occupied by the medial layer (42.97 +/- 5.38%) and was mainly constituted by smooth muscle cells (94.58 +/- 2.65%). All the cell markers used reacted with porcine paraffin-embedded tissue. However, the anti-lysozyme antibody was excluded from this histological analysis because of cytoplasmatic reactivity in smooth muscle cells. In summary, this study proposes histological methods to describe the normal characteristics of the porcine femoral artery and raises the possibility of applying this methodology in future studies on porcine vascular research.
Collapse
Affiliation(s)
- N Solanes
- Institut de Malalties Cardiovasculars, IDIBAPS (Institut d'Investigacions Biomèdiques Agustí Pi Sunyer), Hospital Clínic, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Preclinical restenosis models and drug-eluting stents: still important, still much to learn. J Am Coll Cardiol 2004; 44:1373-85. [PMID: 15464316 DOI: 10.1016/j.jacc.2004.04.060] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2004] [Revised: 03/28/2004] [Accepted: 04/06/2004] [Indexed: 11/30/2022]
Abstract
Percutaneous coronary intervention continues to revolutionize the treatment of coronary atherosclerosis. Restenosis remains a significant problem but may at last be yielding to technologic advances. The examination of neointimal hyperplasia in injured animal artery models has helped in our understanding of angioplasty and stenting mechanisms, and as drug-eluting stent (DES) technologies have arrived, they too have been advanced through the study of animal models. These models are useful for predicting adverse clinical outcomes in patients with DESs because suboptimal animal model studies typically lead to problematic human trials. Similarly, stent thrombosis in animal models suggests stent thrombogenicity in human patients. Equivocal animal model results at six or nine months occasionally have been mirrored by excellent clinical outcomes in patients. The causes of such disparities are unclear but may result from differing methods, including less injury severity than originally described in the models. Ongoing research into animal models will reconcile apparent differences with clinical trials and advance our understanding of how to apply animal models to clinical stenting in the era of DESs.
Collapse
|
43
|
Abstract
Strategies that enhance the number of endothelial cells (ECs) in the vessel wall following injury may limit complications such as thrombosis, vasospasm, and neointimal formation through reconstitution of a luminal barrier and cellular secretion of paracrine factors. Proof of principle has been demonstrated by studies in which mature ECs, culture expanded from harvested vascular tissue, were seeded in the arterial wall following balloon injury. The recent identification of circulating cells capable of developing an endothelial phenotype, including progenitor cells, has raised the possibility of using blood-derived cells as therapeutic agents. This article reviews data suggesting that such cells confer vascular protective effects after injury, raising the potential for novel, autologous approaches to the treatment of vascular disease.
Collapse
Affiliation(s)
- Rajiv Gulati
- Department of Cardiovascular Medicine, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
44
|
Abstract
Restenosis limits the success of percutaneous transluminal coronary interventions. Coronary artery stenting decreases restenosis, improves outcomes, and is currently the most commonly used percutaneous coronary intervention in the United States. However, in-stent restenosis continues to occur at an unacceptable rate. In-stent restenosis is a neointimal hyperplastic response resulting primarily from vascular smooth muscle cell proliferation. Treatment with anti-proliferative agents presents a logical approach to eradicating restenosis, however, these drugs are highly toxic. Coating stents with anti-proliferative agents allows local delivery of high doses and avoids systemic side effects. In 2001, the results of two clinical trials, RAVEL and ELUTES, using sirolimus- and paclitaxil-coated stents demonstrated nearly complete elimination of in-stent restenosis. These dramatic results represent a tremendous advance in the treatment of coronary heart disease.
Collapse
Affiliation(s)
- Dorie W Schwertz
- Department of Medical Surgical Nursing, University of Illinois, Chicago, Illinois, USA
| | | |
Collapse
|
45
|
Affiliation(s)
- W Cwikiel
- Department of Radiology, University of Michigan Hospital, Ann Arbor 48109, USA
| |
Collapse
|
46
|
Abstract
Strategies to treat atherosclerotic coronary artery disease include coronary artery bypass grafting (CABG), in which grafts are used to bypass atherosclerotic vessels and restore blood flow to the ischemic myocardium. The grafts used include healthy arteries or veins harvested from a separate site. Results with arterial grafts have been superior to venous grafts; promoting the practice of total arterial revascularization using only arterial grafts. Suitable arterial grafts, however, are scarce and harvest procedures add to morbidity and cost. Tissue engineering combines the principles of engineering with life sciences for the development of biological substitutes and restore, maintain or improve tissue function. Advances in this field have included the development of tissue-engineered blood vessels, with the potential to serve as arterial grafts, conduits or fistulae. This review describes the history of tissue engineering arteries, the techniques used, and progress to date. The source of cells and the future direction of this field are explored.
Collapse
Affiliation(s)
- Briain D MacNeill
- Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
47
|
Balcells M, Edelman ER. Effect of pre-adsorbed proteins on attachment, proliferation, and function of endothelial cells. J Cell Physiol 2002; 191:155-61. [PMID: 12064458 DOI: 10.1002/jcp.10087] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As certain proteins control cell adhesion, it has been hoped that cell transplantation and tissue engineering could be augmented by pre-adsorption of specific proteins to biological or synthetic surfaces. The questions that remain, however, are whether such proteins can affect cell production as well as adhesion, and if so, whether in a protein-specific manner. We examined the adhesion and the biochemical secretion of bovine aortic endothelial cells (BAEC) on tissue culture polystyrene (TCPS) discs coated with fibronectin (Fn), laminin (Ln), or gelatin. The three coating proteins nonspecifically promote sub-confluent and post-confluent endothelial cell production of total protein up to 2.5-fold of the reference value. Total soluble glycosaminoglycan (GAG) production slightly increased with the different coatings only at low cell density. In contrast, Ln and Fn, not gelatin, drastically enhanced post-confluent BAEC production of prostaglandin (PGI2). However, antibody-blockage of the alpha5 integrin, constituent of the Fn receptor in BAEC, appeared to inhibit the upregulation of PGI2 production observed on Fn-coated surfaces. The results indicate that the cell adhesion mediators used as coating agents dictate cell biological production as well as adhesion and proliferation.
Collapse
Affiliation(s)
- Mercedes Balcells
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, USA.
| | | |
Collapse
|
48
|
Periadventitial Drug Delivery for the Prevention of Restenosis: Rationale and Results. J Vasc Interv Radiol 2002. [DOI: 10.1016/s1051-0443(02)70167-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
49
|
Nugent HM, Edelman ER. Endothelial implants provide long-term control of vascular repair in a porcine model of arterial injury. J Surg Res 2001; 99:228-34. [PMID: 11469891 DOI: 10.1006/jsre.2001.6198] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell culture and animal data support the role of endothelial cells and endothelial-based compounds in regulating vascular repair after injury. We describe a long-term study in pigs in which the biological and immunological responses to endothelial cell implants were investigated 3 months after angioplasty, approximately 2 months after the implants have degraded. Confluent porcine or bovine endothelial cells grown in polymer matrices were implanted adjacent to 28 injured porcine carotid arteries. Porcine and bovine endothelial cell implants significantly reduced experimental restenosis compared to control by 56 and 31%, respectively. Host humoral responses were investigated by detection of an increase in serum antibodies that bind to the bovine or porcine cell strains used for implantation. A significant increase in titer of circulating antibodies to the bovine cells was observed after 4 days in all animals implanted with xenogeneic cells. Detected antibodies returned to presurgery levels after Day 40. No significant increase in titer of antibodies to the porcine cells was observed during the time course of the experiment in animals implanted with porcine endothelial cells. No implanted cells, Gelfoam, or focal inflammatory reaction could be detected histologically at any of the implant sites at 90 days. These data suggest that tissue-engineered endothelial cell implants may provide long-term control of vascular repair after injury, rather than simply delaying lesion formation and that allogeneic implants are able to provide a greater benefit than xenogeneic implants.
Collapse
Affiliation(s)
- H M Nugent
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | | |
Collapse
|
50
|
Gangadharan SP, Eslami MH, Weiss IP, Sui X, Conte MS. Monocyte adhesion to balloon-injured arteries: the influence of endothelial cell seeding. J Vasc Surg 2001; 33:1247-54. [PMID: 11389425 DOI: 10.1067/mva.2001.114211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Deendothelialization of injuries of the artery disrupts normal vascular homeostasis, affecting both the structural integrity of the blood vessel wall, as well as the interaction of the arterial surface with blood components such as platelets, leukocytes, and circulating proteins. Leukocyte and, in particular, monocyte recruitment to damaged vessels has been implicated in the pathogenesis of intimal hyperplasia. We hypothesize that reendothelialization is an important modulator of monocyte adhesion to healing arterial surfaces. METHODS New Zealand white rabbits (n = 20) were subjected to bilateral iliofemoral artery balloon injury. Cultured, autologous venous endothelial cells (ECs) were immediately seeded onto one vessel, whereas the contralateral artery received medium alone, to accelerate endothelial relining. Vessels were harvested (5-9 days after injury) for analysis of permeability (Evans Blue dye exclusion), endothelial coverage (anti-CD31 immunohistochemistry), monocyte adhesion (ex vivo binding of 51Na2CrO4-labeled monocytic THP-1 cells), and monocyte recruitment (RAM-11 immunohistochemistry). RESULTS Improved EC coverage was evidenced by positive staining for CD31 in the seeded vessels. Vessel wall permeability was markedly reduced in EC-seeded arteries (29% +/- 10% vs 99% +/- 0% surface Evans blue staining, P <.005), consistent with restoration of a functional endothelial barrier. EC seeding significantly reduced ex vivo THP-1 binding to vessels explanted at a mean of 8 days after injury (45,170 +/- 8939 vs 85,994 +/- 16,500 cells/cm2, P <.05). However, RAM-11 staining revealed no significant difference in overall macrophage accumulation between seeded and control vessels 1 week after injury (111 +/- 22 vs 95 +/- 14 cells/section, P =.36). CONCLUSIONS Immediate seeding of a balloon-injured rabbit artery with cultured ECs results in accelerated restoration of the endothelial lining. At 1 week, barrier function is improved, and the seeded vessel surface is less adhesive to activated monocytes ex vivo, as compared with injured controls. Nonetheless, EC-seeded and nonseeded arteries demonstrate similar total macrophage accumulation over 1 week. These data suggest that after mechanical arterial injury, endothelial coverage may be one important variable influencing leukocyte adhesion.
Collapse
Affiliation(s)
- S P Gangadharan
- Division of Vascular Surgery, Brigham and Women's Hospital, and the Harvard Institutes of Medicine, Harvard Medical School, Boston, Mass 02115, USA
| | | | | | | | | |
Collapse
|