1
|
Wu M, Sapin-Minet A, Stefan L, Perrin J, Raeth-Fries I, Gaucher C. Heparinized collagen-based hydrogels for tissue engineering: physical, mechanical and biological properties. Int J Pharm 2024; 670:125126. [PMID: 39719257 DOI: 10.1016/j.ijpharm.2024.125126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 12/26/2024]
Abstract
As the main protein forming the vascular extracellular matrix, collagen has a weak antigenicity, making it an attractive candidate for coatings of vascular grafts. In order to bring antithrombotic properties to collagen for obtaining suitable blood compatibility of surfaces and further bioactive molecule carrying capacity, heparinization appears as a method of choice. Thus, in this article, pH-driven self-assembly was used to form collagen-based hydrogels with physical incorporation of heparins, especially low molecular weight heparin or unfractionated heparin at 1 IU/mL and 6 IU/mL. These heparinized hydrogels were evaluated for their physicochemical properties including gelation kinetic, spreading, viscoelasticity, microstructure and heparin quantification, and their biocompatibility such as cytocompatibility and their capacity to release bioactive heparins with antithrombotic properties. The loading capacity of collagen-based hydrogels was higher for unfractionated heparin (60 to 80 %) than for low molecular weight heparin (20 %). Interestingly, the highest concentration (6 IU/mL) of heparin used to form collagen-based hydrogels resulted in the formation of a softer hydrogel owning a better spreadability compared to 1 IU/mL and non heparinized collagen-based hydrogel. The 3D structure observation showed a layered formation with visible pores or spaces between the layers in all types of collagen-based hydrogels. These layers are interconnected by fibrous structures, suggesting a networked architecture. Moreover, the amount of heparin released from collagen-based hydrogel prepared with 6 IU /mL was higher than from those heparinized with 1 IU/mL attested by a delay in blood coagulation (activated partial thromboplastin time and thrombin time) and the abolishment of thrombin generation. Those hydrogels were also biocompatible, with low albumin adsorption and no impact on cell viability. Finally, heparin is retained in these hydrogels for at least seven days after cell seeding, providing the possibility of long-term antithrombotic properties. Thus, we succeeded in develop/obtain coatings with antithrombogenic properties, biocompatible and able to retain heparins at least seven days, making them good candidates with a great potential for the development and application of efficient blood-contacting materials.
Collapse
Affiliation(s)
- Meiling Wu
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | | | - Loïc Stefan
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France
| | - Julien Perrin
- Université de Lorraine, CHRU-Nancy, Service d'hématologie biologique, F-54000 Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | | | | |
Collapse
|
2
|
Song ZP, Chen L, Wang QW, Zhang ZS, Xu JY, Bai WW, Wang SX, Guo T. Circulating monocyte adhesion repairs endothelium-denuded injury through downstream of kinase 3-mediated endothelialization. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167631. [PMID: 39689764 DOI: 10.1016/j.bbadis.2024.167631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
The integrity of the endothelial monolayer is critical for preventing life-threatening hemorrhaging and thrombosis. However, how severe endothelium-denuded injury is rapidly repaired remains unknown. Given the common biological properties between endothelial cells and circulating monocytes, we aimed to examine whether blood monocytes are involved in endothelium wound healing. The in vivo common carotid artery endothelium-denuded (CCAED) model was established through a wire-induced injury. Monocyte adhesion was assessed using immunofluorescence and a parallel plate flow chamber. We initially observed that the circulating monocyte-mediated endothelialization was better downstream of kinase 3 deficient mice (DOK3-/-) than that of wild-type (WT) mice following induction of the CCAED model. Rapid endothelialization increased endothelial integrity, prevented coagulation, and decreased thrombosis. Mechanistically, following endothelium-denuded injury, monocyte chemoattractant protein 1 (MCP1) disassociated from DOK3 and C-C chemokine receptor type 2B (CCR2B), increased the intracellular Ca2+ concentration, and promoted adhesion in circulating monocytes. However, this process was inhibited by the CCR2B inhibitor INCB3344. Moreover, the adhesive functions of circulating monocytes isolated from DOK3-/- mice were stronger than those from WT mice. Furthermore, adhered monocytes expressed endothelial-specific markers and compensated for endothelium-dependent vasorelaxation in WT mice. Similarly, these effects were enhanced in DOK3-/- mice. Bindarit, a selective MCP1 inhibitor, suppressed endothelialization following CCAED surgery in WT mice but not in DOK3-/- mice. In conclusion, endothelialization mediated by circulating monocytes repairs endothelium-denuded injury to compensate for endothelial functions through MCP1/DOK3/CCR2B/Ca2+ signaling. Our findings indicate that circulating monocyte adhesion is an important endothelial wound healing mechanism.
Collapse
Affiliation(s)
- Zhi-Peng Song
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Chen
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qian-Wen Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhen-Shan Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jia-Yao Xu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wen-Wu Bai
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Shuang-Xi Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Tao Guo
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Snyder Y, Jana S. Innovative Substrate Design with Basement Membrane Components for Enhanced Endothelial Cell Function and Endothelization. Adv Healthc Mater 2024; 13:e2401150. [PMID: 39021293 DOI: 10.1002/adhm.202401150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/28/2024] [Indexed: 07/20/2024]
Abstract
Enhancing endothelial cell growth on small-diameter vascular grafts produced from decellularized tissues or synthetic substrates is pivotal for preventing thrombosis. While optimized decellularization protocols can preserve the structure and many components of the extracellular matrix (ECM), the process can still lead to the loss of crucial basement membrane proteins, such as laminin, collagen IV, and perlecan, which are pivotal for endothelial cell adherence and functional growth. This loss can result in poor endothelialization and endothelial cell activation causing thrombosis and intimal hyperplasia. To address this, the basement membrane's ECM is emulated on fiber substrates, providing a more physiological environment for endothelial cells. Thus, fibroblasts are cultured on fiber substrates to produce an ECM membrane substrate (EMMS) with basement membrane proteins. The EMMS then underwent antigen removal (AR) treatment to eliminate antigens from the membrane while preserving essential proteins and producing an AR-treated membrane substrate (AMS). Subsequently, human endothelial cells cultured on the AMS exhibited superior proliferation, nitric oxide production, and increased expression of endothelial markers of quiescence/homeostasis, along with autophagy and antithrombotic factors, compared to those on the decellularized aortic tissue. This strategy showed the potential of pre-endowing fiber substrates with a basement membrane to enable better endothelization.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Chemical and Biomedical Engineering, University of Missouri, 1406 Rollins Street, Columbia, MO, 65211, USA
| | - Soumen Jana
- Department of Chemical and Biomedical Engineering, University of Missouri, 1406 Rollins Street, Columbia, MO, 65211, USA
| |
Collapse
|
4
|
Lorentz KL, Marini AX, Bruk LA, Gupta P, Mandal BB, DiLeo MV, Weinbaum JS, Little SR, Vorp DA. Mesenchymal Stem Cell-Conditioned Media-Loaded Microparticles Enhance Acute Patency in Silk-Based Vascular Grafts. Bioengineering (Basel) 2024; 11:947. [PMID: 39329689 PMCID: PMC11428691 DOI: 10.3390/bioengineering11090947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/10/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Coronary artery disease leads to over 360,000 deaths annually in the United States, and off-the-shelf bypass graft options are currently limited and/or have high failure rates. Tissue-engineered vascular grafts (TEVGs) present an attractive option, though the promising mesenchymal stem cell (MSC)-based implants face uncertain regulatory pathways. In this study, "artificial MSCs" (ArtMSCs) were fabricated by encapsulating MSC-conditioned media (CM) in poly(lactic-co-glycolic acid) microparticles. ArtMSCs and control microparticles (Blank-MPs) were incubated over 7 days to assess the release of total protein and the vascular endothelial growth factor (VEGF-A); releasates were also assessed for cytotoxicity and promotion of smooth muscle cell (SMC) proliferation. Each MP type was loaded in previously published "lyogel" silk scaffolds and implanted as interposition grafts in Lewis rats for 1 or 8 weeks. Explanted grafts were assessed for patency and cell content. ArtMSCs had a burst release of protein and VEGF-A. CM increased proliferation in SMCs, but not after encapsulation. TEVG explants after 1 week had significantly higher patency rates with ArtMSCs compared to Blank-MPs, but similar to unseeded lyogel grafts. ArtMSC explants had lower numbers of infiltrating macrophages compared to Blank-MP explants, suggesting a modulation of inflammatory response by the ArtMSCs. TEVG explants after 8 weeks showed no significant difference in patency among the three groups. The ArtMSC explants showed higher numbers of SMCs and endothelial cells within the neotissue layer of the graft compared to Blank-MP explants. In sum, while the ArtMSCs had positive effects acutely, efficacy was lost in the longer term; therefore, further optimization is needed.
Collapse
Affiliation(s)
- Katherine L Lorentz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ande X Marini
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Liza A Bruk
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Prerak Gupta
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Morgan V DiLeo
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Steven R Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, PA 15261, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee Women's Research Institute, Pittsburgh, PA 15213, USA
| |
Collapse
|
5
|
DeMaria WG, Figueroa-Milla AE, Kaija A, Harrington AE, Tero B, Ryzhova L, Liaw L, Rolle MW. Endothelial Cells Increase Mesenchymal Stem Cell Differentiation in Scaffold-Free 3D Vascular Tissue. Tissue Eng Part A 2024. [PMID: 39109944 DOI: 10.1089/ten.tea.2024.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
In this study, we present a versatile, scaffold-free approach to create ring-shaped engineered vascular tissue segments using human mesenchymal stem cell-derived smooth muscle cells (hMSC-SMCs) and endothelial cells (ECs). We hypothesized that incorporation of ECs would increase hMSC-SMC differentiation without compromising tissue ring strength or fusion to form tissue tubes. Undifferentiated hMSCs and ECs were co-seeded into custom ring-shaped agarose wells using four different concentrations of ECs: 0%, 10%, 20%, and 30%. Co-seeded EC and hMSC rings were cultured in SMC differentiation medium for a total of 22 days. Tissue rings were then harvested for histology, Western blotting, wire myography, and uniaxial tensile testing to examine their structural and functional properties. Differentiated hMSC tissue rings comprising 20% and 30% ECs exhibited significantly greater SMC contractile protein expression, endothelin-1 (ET-1)-meditated contraction, and force at failure compared with the 0% EC rings. On average, the 0%, 10%, 20%, and 30% EC rings exhibited a contractile force of 0.745 ± 0.117, 0.830 ± 0.358, 1.31 ± 0.353, and 1.67 ± 0.351 mN (mean ± standard deviation [SD]) in response to ET-1, respectively. Additionally, the mean maximum force at failure for the 0%, 10%, 20%, and 30% EC rings was 88.5 ± 36. , 121 ± 59.1, 147 ± 43.1, and 206 ± 0.8 mN (mean ± SD), respectively. Based on these results, 30% EC rings were fused together to form tissue-engineered blood vessels (TEBVs) and compared with 0% EC TEBV controls. The addition of 30% ECs in TEBVs did not affect ring fusion but did result in significantly greater SMC protein expression (calponin and smoothelin). In summary, co-seeding hMSCs with ECs to form tissue rings resulted in greater contraction, strength, and hMSC-SMC differentiation compared with hMSCs alone and indicates a method to create a functional 3D human vascular cell coculture model.
Collapse
Affiliation(s)
- William G DeMaria
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Andre E Figueroa-Milla
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Abigail Kaija
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | | | - Benjamin Tero
- MaineHealth Institute for Research, Scarborough, Maine, USA
- The Roux Institute, Northeastern University, Portland, Maine, USA
| | - Larisa Ryzhova
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | - Lucy Liaw
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | - Marsha W Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- The Roux Institute, Northeastern University, Portland, Maine, USA
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Jiang H, Sun X, Wu Y, Xu J, Xiao C, Liu Q, Fang L, Liang Y, Zhou J, Wu Y, Lin Z. Contribution of Tregs to the promotion of constructive remodeling after decellularized extracellular matrix material implantation. Mater Today Bio 2024; 27:101151. [PMID: 39104900 PMCID: PMC11298607 DOI: 10.1016/j.mtbio.2024.101151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 08/07/2024] Open
Abstract
Host remodeling of decellularized extracellular matrix (dECM) material through the appropriate involvement of immune cells is essential for achieving functional organ/tissue regeneration. As many studies have focused on the role of macrophages, only few have evaluated the role of regulatory T cells (Tregs) in dECM remodeling. In this study, we used a mouse model of traumatic muscle injury to determine the role of Tregs in the constructive remodeling of vascular-derived dECM. According to the results, a certain number of Tregs could be recruited after dECM implantation. Notably, using anti-CD25 to reduce the number of Tregs recruited by the dECM was significantly detrimental to material remodeling based on a significant reduction in the number of M2 macrophages. In addition, collagen and elastic fibers, which maintain the integrity and mechanical properties of the material, rapidly degraded during the early stages of implantation. In contrast, the use of CD28-SA antibodies to increase the number of Tregs recruited by dECM promoted constructive remodeling, resulting in a decreased inflammatory response at the material edge, thinning of the surrounding fibrous connective tissue, uniform infiltration of host cells, and significantly improved tissue remodeling scores. The number of M2 macrophages increased whereas that of M1 macrophages decreased. Moreover, Treg-conditioned medium further enhanced material-induced M2 macrophage polarization in vitro. Overall, Treg is an important cell type that influences constructive remodeling of the dECM. Such findings contribute to the design of next-generation biomaterials to optimize the remodeling and regeneration of dECM materials.
Collapse
Affiliation(s)
- Hongjing Jiang
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
| | - Xuheng Sun
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Yindi Wu
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
| | - Jianyi Xu
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Cong Xiao
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Qing Liu
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Lijun Fang
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Yuanfeng Liang
- Department of Geriatrics, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510006, Guangzhou, Guangdong, China
| | - Jiahui Zhou
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
| | - Yueheng Wu
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, 528200, Foshan, Guangdong, China
| | - Zhanyi Lin
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, 528200, Foshan, Guangdong, China
| |
Collapse
|
7
|
Ding Y, Warlick L, Chen M, Taddese E, Collins C, Fu R, Duan C, Wang X, Ware H, Sun C, Ameer G. 3D-printed, citrate-based bioresorbable vascular scaffolds for coronary artery angioplasty. Bioact Mater 2024; 38:195-206. [PMID: 38756202 PMCID: PMC11096684 DOI: 10.1016/j.bioactmat.2024.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/18/2024] Open
Abstract
Fully bioresorbable vascular scaffolds (BVSs) aim to overcome the limitations of metallic drug-eluting stents (DESs). However, polymer-based BVSs, such as Abbott's Absorb, the only US FDA-approved BVS, have had limited use due to increased strut thickness (157 μm for Absorb), exacerbated tissue inflammation, and increased risk of major cardiac events leading to inferior clinical performance when compared to metallic DESs. Herein we report the development of a drug-eluting BVS (DE-BVS) through the innovative use of a photopolymerizable, citrate-based biomaterial and a high-precision additive manufacturing process. BVS with a clinically relevant strut thickness of 62 μm can be produced in a high-throughput manner, i.e. one BVS per minute, and controlled release of the anti-restenosis drug everolimus can be achieved by engineering the structure of polymer coatings to fabricate drug-eluting BVS. We achieved the successful deployment of BVSs and DE-BVSs in swine coronary arteries using a custom-built balloon catheter and BVS delivery system and confirmed BVS safety and efficacy regarding maintenance of vessel patency for 28 days, observing an inflammation profile for BVS and DE-BVS that was comparable to the commercial XIENCE™ DES (Abbott Vascular).
Collapse
Affiliation(s)
- Yonghui Ding
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Liam Warlick
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Mian Chen
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Eden Taddese
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Caralyn Collins
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Rao Fu
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Chongwen Duan
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Xinlong Wang
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Henry Ware
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Cheng Sun
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Guillermo Ameer
- Centre for Advanced Regenerative Engineering (CARE), Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
8
|
Das A, Smith RJ, Andreadis ST. Harnessing the potential of monocytes/macrophages to regenerate tissue-engineered vascular grafts. Cardiovasc Res 2024; 120:839-854. [PMID: 38742656 PMCID: PMC11218695 DOI: 10.1093/cvr/cvae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
| | - Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, 701 Ellicott St, Buffalo, NY 14203, USA
- Cell, Gene and Tissue Engineering (CGTE) Center, University at Buffalo, The State University of New York, 813 Furnas Hall, Buffalo, NY 14260-4200, USA
| |
Collapse
|
9
|
Las Heras K, Garcia-Orue I, Rancan F, Igartua M, Santos-Vizcaino E, Hernandez RM. Modulating the immune system towards a functional chronic wound healing: A biomaterials and Nanomedicine perspective. Adv Drug Deliv Rev 2024; 210:115342. [PMID: 38797316 DOI: 10.1016/j.addr.2024.115342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Chronic non-healing wounds persist as a substantial burden for healthcare systems, influenced by factors such as aging, diabetes, and obesity. In contrast to the traditionally pro-regenerative emphasis of therapies, the recognition of the immune system integral role in wound healing has significantly grown, instigating an approach shift towards immunological processes. Thus, this review explores the wound healing process, highlighting the engagement of the immune system, and delving into the behaviors of innate and adaptive immune cells in chronic wound scenarios. Moreover, the article investigates biomaterial-based strategies for the modulation of the immune system, elucidating how the adjustment of their physicochemical properties or their synergistic combination with other agents such as drugs, proteins or mesenchymal stromal cells can effectively modulate the behaviors of different immune cells. Finally this review explores various strategies based on synthetic and biological nanostructures, including extracellular vesicles, to finely tune the immune system as natural immunomodulators or therapeutic nanocarriers with promising biophysical properties.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itxaso Garcia-Orue
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Fiorenza Rancan
- Department of Dermatology, Venereology und Allergology,Clinical Research Center for Hair and Skin Science, Charité - Universitätsmedizin Berlin
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Wang Z, Zhang M, Liu L, Mithieux SM, Weiss AS. Polyglycerol sebacate-based elastomeric materials for arterial regeneration. J Biomed Mater Res A 2024; 112:574-585. [PMID: 37345954 DOI: 10.1002/jbm.a.37583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/15/2023] [Accepted: 06/07/2023] [Indexed: 06/23/2023]
Abstract
Synthetic vascular grafts are commonly used in patients with severe occlusive arterial disease when autologous grafts are not an option. Commercially available synthetic grafts are confronted with challenging outcomes: they have a lower patency rate than autologous grafts and are currently unable to promote arterial regeneration. Polyglycerol sebacate (PGS), a non-toxic polymer with a tunable degradation profile, has shown promising results as a small-diameter vascular graft component that can support the formation of neoarteries. In this review, we first present an overview of the synthesis and modification of PGS followed by an examination of its mechanical properties. We then report on the performance, degradation, regeneration, and remodeling of PGS-based small-diameter vascular grafts, with a focus on efforts to reduce thrombosis, prevent dilation, and promote cellular residency and extracellular matrix regeneration that resembles the native artery in spatial distribution and organization. We also highlight recent advances in the incorporation of novel in situ cell sources for arterial regeneration and their potential application in PGS-based vascular grafts. Finally, we compare vascular grafts fabricated using PGS-based materials with other elastomeric alternatives.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Miao Zhang
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Linyang Liu
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Suzanne M Mithieux
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony S Weiss
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- The University of Sydney Nano Institute, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
11
|
Fahad MAA, Lee HY, Park S, Choi M, Shanto PC, Park M, Bae SH, Lee BT. Small-diameter vascular graft composing of core-shell structured micro-nanofibers loaded with heparin and VEGF for endothelialization and prevention of neointimal hyperplasia. Biomaterials 2024; 306:122507. [PMID: 38367300 DOI: 10.1016/j.biomaterials.2024.122507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
Despite the significant progress made in recent years, clinical issues with small-diameter vascular grafts related to low mechanical strength, thrombosis, intimal hyperplasia, and insufficient endothelialization remain unresolved. This study aims to design and fabricate a core-shell fibrous small-diameter vascular graft by co-axial electrospinning process, which will mechanically and biologically meet the benchmarks for blood vessel replacement. The presented graft (PGHV) comprised polycaprolactone/gelatin (shell) loaded with heparin-VEGF and polycaprolactone (core). This study hypothesized that the shell structure of the fibers would allow rapid degradation to release heparin-VEGF, and the core would provide mechanical strength for long-term application. Physico-mechanical evaluation, in vitro biocompatibility, and hemocompatibility assays were performed to ensure safe in vivo applications. After 25 days, the PGHV group released 79.47 ± 1.54% of heparin and 86.25 ± 1.19% of VEGF, and degradation of the shell was observed but the core remained pristine. Both the control (PG) and PGHV groups demonstrated robust mechanical properties. The PGHV group showed excellent biocompatibility and hemocompatibility compared to the PG group. After four months of rat aorta implantation, PGHV exhibited smooth muscle cell regeneration and complete endothelialization with a patency rate of 100%. The novel core-shell structured graft could be pivotal in vascular tissue regeneration application.
Collapse
Affiliation(s)
- Md Abdullah Al Fahad
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hyun-Yong Lee
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, 31151, Republic of Korea
| | - Seongsu Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Minji Choi
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Prayas Chakma Shanto
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Sang Ho Bae
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, 31151, Republic of Korea; Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, 31151, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea; Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| |
Collapse
|
12
|
Paone L, Szkolnicki M, DeOre BJ, Tran KA, Goldman N, Andrews AM, Ramirez SH, Galie PA. Effects of Drag-Reducing Polymers on Hemodynamics and Whole Blood-Endothelial Interactions in 3D-Printed Vascular Topologies. ACS APPLIED MATERIALS & INTERFACES 2024; 16:14457-14466. [PMID: 38488736 PMCID: PMC10982934 DOI: 10.1021/acsami.3c17099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Most in vitro models use culture medium to apply fluid shear stress to endothelial cells, which does not capture the interaction between blood and endothelial cells. Here, we describe a new system to characterize whole blood flow through a 3D-printed, endothelialized vascular topology that induces flow separation at a bifurcation. Drag-reducing polymers, which have been previously studied as a potential therapy to reduce the pressure drop across the vascular bed, are evaluated for their effect on mitigating the disturbed flow. Polymer concentrations of 1000 ppm prevented recirculation and disturbed flow at the wall. Proteomic analysis of plasma collected from whole blood recirculated through the vascularized channel with and without drag-reducing polymers provides insight into the effects of flow regimes on levels of proteins indicative of the endothelial-blood interaction. The results indicate that blood flow alters proteins associated with coagulation, inflammation, and other processes. Overall, these proof-of-concept experiments demonstrate the importance of using whole blood flow to study the endothelial response to perfusion.
Collapse
Affiliation(s)
- Louis
S. Paone
- Department
of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Matthew Szkolnicki
- Department
of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Brandon J. DeOre
- Department
of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Kiet A. Tran
- Department
of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Noah Goldman
- Department
of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Allison M. Andrews
- Department
of Pathology, Immunology, & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Servio H. Ramirez
- Department
of Pathology, Immunology, & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Peter A. Galie
- Department
of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
13
|
Wu H, Yang L, Luo R, Li L, Zheng T, Huang K, Qin Y, Yang X, Zhang X, Wang Y. A drug-free cardiovascular stent functionalized with tailored collagen supports in-situ healing of vascular tissues. Nat Commun 2024; 15:735. [PMID: 38272886 PMCID: PMC10810808 DOI: 10.1038/s41467-024-44902-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
Drug-eluting stent implantation suppresses the excessive proliferation of smooth muscle cells to reduce in-stent restenosis. However, the efficacy of drug-eluting stents remains limited due to delayed reendothelialization, impaired intimal remodeling, and potentially increased late restenosis. Here, we show that a drug-free coating formulation functionalized with tailored recombinant humanized type III collagen exerts one-produces-multi effects in response to injured tissue following stent implantation. We demonstrate that the one-produces-multi coating possesses anticoagulation, anti-inflammatory, and intimal hyperplasia suppression properties. We perform transcriptome analysis to indicate that the drug-free coating favors the endothelialization process and induces the conversion of smooth muscle cells to a contractile phenotype. We find that compared to drug-eluting stents, our drug-free stent reduces in-stent restenosis in rabbit and porcine models and improves vascular neointimal healing in a rabbit model. Collectively, the one-produces-multi drug-free system represents a promising strategy for the next-generation of stents.
Collapse
Affiliation(s)
- Haoshuang Wu
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Tiantian Zheng
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Kaiyang Huang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yumei Qin
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Xia Yang
- Shanxi Key Laboratory of Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd., Taiyuan, 030032, Shanxi, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China.
- Tianfu Jincheng Laboratory (Frontier Medical Center), Chengdu, 610213, China.
| |
Collapse
|
14
|
Azman SS, Yazid MD, Abdul Ghani NA, Raja Sabudin RZA, Abdul Rahman MR, Sulaiman N. Generation of a novel ex-vivo model to study re-endothelialization. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:408-416. [PMID: 37584645 DOI: 10.1080/21691401.2023.2245456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
Endothelial dysfunction initiates the pathogenesis of a myriad of cardiovascular diseases, yet the precise underlying mechanisms remain unclear. Current model utilises mechanical denudation of arteries resulting in an arterial-injury model with onset of intimal hyperplasia (IH). Our study shows that 5 min enzymatic denudation of human umbilical artery (hUA) lumen at 37 °C efficiently denudes hUA while maintaining vessel integrity without significantly increase intima-media thickness after 7 days in culture. This ex-vivo model will be a valuable tool in understanding the mechanism of re-endothelialization prior to smooth muscle cells (SMC) activation thus placating IH at an early stage.
Collapse
Affiliation(s)
- Siti Sarah Azman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Faculty of Applied Sciences, Universiti Teknologi MARA, Perak Branch, Tapah Campus, Perak, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nur Azurah Abdul Ghani
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Hospital Canselor Tuanku Mukhriz, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Raja Zahratul Azma Raja Sabudin
- Hospital Canselor Tuanku Mukhriz, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Hospital Canselor Tuanku Mukhriz, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| |
Collapse
|
15
|
Mahara A, Shirai M, Soni R, Le HT, Shimizu K, Hirano Y, Yamaoka T. Vascular tissue reconstruction by monocyte subpopulations on small-diameter acellular grafts via integrin activation. Mater Today Bio 2023; 23:100847. [PMID: 37953756 PMCID: PMC10632538 DOI: 10.1016/j.mtbio.2023.100847] [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: 06/29/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Although the clinical application of cell-free tissue-engineered vascular grafts (TEVGs) has been proposed, vascular tissue regeneration mechanisms have not been fully clarified. Here, we report that monocyte subpopulations reconstruct vascular-like tissues through integrin signaling. An Arg-Glu-Asp-Val peptide-modified acellular long-bypass graft was used as the TEVG, and tissue regeneration in the graft was evaluated using a cardiopulmonary pump system and porcine transplantation model. In 1 day, the luminal surface of the graft was covered with cells that expressed CD163, CD14, and CD16, which represented the monocyte subpopulation, and they exhibited proliferative and migratory abilities. RNA sequencing showed that captured cells had an immune-related phenotype similar to that of monocytes and strongly expressed cell adhesion-related genes. In vitro angiogenesis assay showed that tube formation of the captured cells occurred via integrin signal activation. After medium- and long-term graft transplantation, the captured cells infiltrated the tunica media layer and constructed vascular with a CD31/CD105-positive layer and an αSMA-positive structure after 3 months. This finding, including multiple early-time observations provides clear evidence that blood-circulating monocytes are directly involved in vascular remodeling.
Collapse
Affiliation(s)
- Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Manabu Shirai
- Omics Research Center, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Raghav Soni
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Hue Thi Le
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Kaito Shimizu
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka, 565-8680, Japan
| | - Yoshiaki Hirano
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka, 565-8680, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| |
Collapse
|
16
|
Yang T, Li G, Li X, Wei B, Su H, Liu W, Guo S, Yang N, Xu T, Duan C. VEGF combined with DAPT promotes tissue regeneration and remodeling in vascular grafts. Regen Biomater 2023; 10:rbad088. [PMID: 37899954 PMCID: PMC10603585 DOI: 10.1093/rb/rbad088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 10/31/2023] Open
Abstract
Previous research on tissue-engineered blood vessels (TEBVs) has mainly focused on the intima or adventitia unilaterally, neglecting the equal importance of both layers. Meanwhile, the efficacy of grafts modified with vascular endothelial growth factor (VEGF) merely has been limited. Here, we developed a small-diameter graft that can gradually release VEGF and γ secretase inhibitor IX (DAPT) to enhance tissue regeneration and remodeling in both the intima and adventitia. In vitro, experiments revealed that the combination of VEGF and DAPT had superior pro-proliferation and pro-migration effects on endothelial cells. In vivo, the sustained release of VEGF and DAPT from the grafts resulted in improved regeneration and remodeling. Specifically, in the intima, faster endothelialization and regeneration of smooth muscle cells led to higher patency rates and better remodeling. In the adventitia, a higher density of neovascularization, M2 macrophages and fibroblasts promoted cellular ingrowth and replacement of the implant with autologous neo-tissue. Furthermore, western blot analysis confirmed that the regenerated ECs were functional and the effect of DAPT was associated with increased expression of vascular endothelial growth factor receptor 2. Our study demonstrated that the sustained release of VEGF and DAPT from the graft can effectively promote tissue regeneration and remodeling in both the intima and adventitia. This development has the potential to significantly accelerate the clinical application of small-diameter TEBVs.
Collapse
Affiliation(s)
- Tao Yang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Guangxu Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Xifeng Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Boyang Wei
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hengxian Su
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wenchao Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Shenquan Guo
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Nan Yang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Xu
- Department of Bio-intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen 518057, People’s Republic of China
- Department of Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, People’s Republic of China
| | - Chuanzhi Duan
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
17
|
Hao H, Xue Y, Wu Y, Wang C, Chen Y, Wang X, Zhang P, Ji J. A paradigm for high-throughput screening of cell-selective surfaces coupling orthogonal gradients and machine learning-based cell recognition. Bioact Mater 2023; 28:1-11. [PMID: 37214260 PMCID: PMC10192934 DOI: 10.1016/j.bioactmat.2023.04.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
The combinational density of immobilized functional molecules on biomaterial surfaces directs cell behaviors. However, limited by the low efficiency of traditional low-throughput experimental methods, investigation and optimization of the combinational density remain daunting challenges. Herein, we report a high-throughput screening set-up to study biomaterial surface functionalization by integrating photo-controlled thiol-ene surface chemistry and machine learning-based label-free cell identification and statistics. Through such a strategy, a specific surface combinational density of polyethylene glycol (PEG) and arginine-glutamic acid-aspartic acid-valine peptide (REDV) leads to high endothelial cell (EC) selectivity against smooth muscle cell (SMC) was identified. The composition was translated as a coating formula to modify medical nickel-titanium alloy surfaces, which was then proved to improve EC competitiveness and induce endothelialization. This work provided a high-throughput method to investigate behaviors of co-cultured cells on biomaterial surfaces modified with combinatorial functional molecules.
Collapse
Affiliation(s)
- Hongye Hao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Yunfan Xue
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Yuhui Wu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Cong Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Yifeng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Xingwang Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, 314400, PR China
| |
Collapse
|
18
|
Ibrahim DM, Fomina A, Bouten CVC, Smits AIPM. Functional regeneration at the blood-biomaterial interface. Adv Drug Deliv Rev 2023; 201:115085. [PMID: 37690484 DOI: 10.1016/j.addr.2023.115085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/01/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
The use of cardiovascular implants is commonplace in clinical practice. However, reproducing the key bioactive and adaptive properties of native cardiovascular tissues with an artificial replacement is highly challenging. Exciting new treatment strategies are under development to regenerate (parts of) cardiovascular tissues directly in situ using immunomodulatory biomaterials. Direct exposure to the bloodstream and hemodynamic loads is a particular challenge, given the risk of thrombosis and adverse remodeling that it brings. However, the blood is also a source of (immune) cells and proteins that dominantly contribute to functional tissue regeneration. This review explores the potential of the blood as a source for the complete or partial in situ regeneration of cardiovascular tissues, with a particular focus on the endothelium, being the natural blood-tissue barrier. We pinpoint the current scientific challenges to enable rational engineering and testing of blood-contacting implants to leverage the regenerative potential of the blood.
Collapse
Affiliation(s)
- Dina M Ibrahim
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Aleksandra Fomina
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Graduate School of Life Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
19
|
Changizi S, Sameti M, Bazemore GL, Chen H, Bashur CA. Epsin Mimetic UPI Peptide Delivery Strategies to Improve Endothelization of Vascular Grafts. Macromol Biosci 2023; 23:e2300073. [PMID: 37117010 DOI: 10.1002/mabi.202300073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Indexed: 04/30/2023]
Abstract
Endothelialization of engineered vascular grafts for replacement of small-diameter coronary arteries remains a critical challenge. The ability for an acellular vascular graft to promote endothelial cell (EC) recruitment in the body would be very beneficial. This study investigated epsins as a target since they are involved in internalization of vascular endothelial growth factor receptor 2. Specifically, epsin-mimetic UPI peptides are delivered locally from vascular grafts to block epsin activity and promote endothelialization. The peptide delivery from fibrin coatings allowed for controlled loading and provided a significant improvement in EC attachment, migration, and growth in vitro. The peptides have even more important impacts after grafting into rat abdominal aortae. The peptides prevented graft thrombosis and failure that is observed with a fibrin coating alone. They also modulated the in vivo remodeling. The grafts are able to remodel without the formation of a thick fibrous capsule on the adventitia with the 100 µg mL-1 peptide-loaded condition, and this condition enabled the formation of a functional EC monolayer in the graft lumen after only 1 week. Overall, this study demonstrated that the local delivery of UPI peptides is a promising strategy to improve the performance of vascular grafts.
Collapse
Affiliation(s)
- Shirin Changizi
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, 32901, USA
| | - Mahyar Sameti
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, 32901, USA
| | - Gabrielle L Bazemore
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, 32901, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Chris A Bashur
- Department of Biomedical Engineering, Florida Institute of Technology, Melbourne, FL, 32901, USA
| |
Collapse
|
20
|
Huang LP, Liu Y, Li QJ, Zhang WQ, Wu CY, Zhao LM, Xie HQ. A Modified Small Intestinal Submucosa Patch with Multifunction to Promote Scarless Repair and Reinvigoration of Urethra. Adv Healthc Mater 2023; 12:e2300519. [PMID: 37062917 DOI: 10.1002/adhm.202300519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Indexed: 04/18/2023]
Abstract
To reconstruct and restore the functions of the male urethra is a challenging task for urologists. The acellular matrix graft currently used in the clinics is mono-functional and may cause a series of complications including stricture, fibrosis, and stone formation. As a result, such graft materials cannot meet the increasing demand for multifunctionality in the field of urethral tissue engineering. In this context, a multifunctional urethral patch is designed for the repair of urethral defects by mixing protocatechualdehyde (PCA) with small intestinal submucosa (SIS) under an alkalin condition to allow cross linking. As shown, the PCA/SIS patch possesses excellent biocompatibility, antioxidant activity, and anti-inflammatory property. More importantly, this patch can remarkably promote the adhesion, proliferation, and directional extension of rabbit bladder epithelial mucous cells (R-EMCs) as well as rabbit bladder smooth muscle cells (R-SMCs), and upregulate the expression of cytokeratin in the EMCs and contractile protein in the SMCs in vitro. In vivo experiments also confirm that the PCA/SIS patch can significantly enhance scarless repair of urethral defects in rabbits by facilitating smooth muscle regeneration, reducing excessive collagen deposition, and accelerating re-epithelialization and neovascularization. Taken together, the newly developed multifunctional PCA/SIS patch provides a promising candidate for urethral regeneration.
Collapse
Affiliation(s)
- Li-Ping Huang
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuan Liu
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qian-Jin Li
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wen-Qian Zhang
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chen-Yu Wu
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Long-Mei Zhao
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hui-Qi Xie
- Department of Orthopedics Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
21
|
Tao H, Zhong X, Zeng A, Song L. Unveiling the veil of lactate in tumor-associated macrophages: a successful strategy for immunometabolic therapy. Front Immunol 2023; 14:1208870. [PMID: 37564659 PMCID: PMC10411982 DOI: 10.3389/fimmu.2023.1208870] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
Lactate, traditionally regarded as a metabolic waste product at the terminal of the glycolysis process, has recently been found to have multifaceted functional roles in metabolism and beyond. A metabolic reprogramming phenomenon commonly seen in tumor cells, known as the "Warburg effect," sees high levels of aerobic glycolysis result in an excessive production of lactate. This lactate serves as a substrate that sustains not only the survival of cancer cells but also immune cells. However, it also inhibits the function of tumor-associated macrophages (TAMs), a group of innate immune cells ubiquitously present in solid tumors, thereby facilitating the immune evasion of malignant tumor cells. Characterized by their high plasticity, TAMs are generally divided into the pro-inflammatory M1 phenotype and the pro-tumour M2 phenotype. Through a process of 'education' by lactate, TAMs tend to adopt an immunosuppressive phenotype and collaborate with tumor cells to promote angiogenesis. Additionally, there is growing evidence linking metabolic reprogramming with epigenetic modifications, suggesting the participation of histone modification in diverse cellular events within the tumor microenvironment (TME). In this review, we delve into recent discoveries concerning lactate metabolism in tumors, with a particular focus on the impact of lactate on the function of TAMs. We aim to consolidate the molecular mechanisms underlying lactate-induced TAM polarization and angiogenesis and explore the lactate-mediated crosstalk between TAMs and tumor cells. Finally, we also touch upon the latest progress in immunometabolic therapies and drug delivery strategies targeting glycolysis and lactate production, offering new perspectives for future therapeutic approaches.
Collapse
Affiliation(s)
- Hongxia Tao
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xuansheng Zhong
- Clinical Medicine Department, Bengbu Medical College, Bengbu, China
| | - Anqi Zeng
- Institute of Translational Pharmacology and Clinical Application, Sichuan Academy of Chinese Medical Science, Chengdu, Sichuan, China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
22
|
Yao Y, Zaw AM, Anderson DE, Jeong Y, Kunihiro J, Hinds MT, Yim EK. Fucoidan and topography modification improved in situ endothelialization on acellular synthetic vascular grafts. Bioact Mater 2023; 22:535-550. [PMID: 36330164 PMCID: PMC9619221 DOI: 10.1016/j.bioactmat.2022.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/20/2022] [Accepted: 10/09/2022] [Indexed: 11/13/2022] Open
Abstract
Thrombogenesis remains the primary failure of synthetic vascular grafts. Endothelial coverage is crucial to provide an antithrombogenic surface. However, most synthetic materials do not support cell adhesion, and transanastomotic endothelial migration is limited. Here, a surface modification strategy using fucoidan and topography was developed to enable fast in situ endothelialization of polyvinyl alcohol, which is not endothelial cell-adhesive. Among three different immobilization approaches compared, conjugation of aminated-fucoidan promoted endothelial monolayer formation while minimizing thrombogenicity in both in vitro platelet rich plasma testing and ex vivo non-human primate shunt assay. Screening of six topographical patterns showed that 2 μm gratings increased endothelial cell migration without inducing inflammation responses of endothelial cells. Mechanistic studies demonstrated that fucoidan could attract fibronectin, enabling integrin binding and focal adhesion formation and activating focal adhesion kinase (FAK) signaling, and 2 μm gratings further enhanced FAK-mediated cell migration. In a clinically relevant rabbit carotid artery end-to-side anastomosis model, 60% in situ endothelialization was observed throughout the entire lumen of 1.7 mm inner diameter modified grafts, compared to 0% of unmodified graft, and the four-week graft patency also increased. This work presents a promising strategy to stimulate in situ endothelialization on synthetic materials for improving long-term performance.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Aung Moe Zaw
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Deirdre E.J. Anderson
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - YeJin Jeong
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Joshua Kunihiro
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Evelyn K.F. Yim
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Center for Biotechnology and Bioengineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
23
|
Wu Y, Yazdani SK, Bolander JEM, Wagner WD. Syndecan-4 and stromal cell-derived factor-1 alpha functionalized endovascular scaffold facilitates adhesion, spreading and differentiation of endothelial colony forming cells and functions under flow and shear stress conditions. J Biomed Mater Res B Appl Biomater 2023; 111:538-550. [PMID: 36208170 PMCID: PMC10092721 DOI: 10.1002/jbm.b.35170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/22/2022] [Accepted: 09/03/2022] [Indexed: 01/25/2023]
Abstract
Acellular vascular scaffolds with capture molecules have shown great promise in recruiting circulating endothelial colony forming cells (ECFCs) to promote in vivo endothelialization. A microenvironment conducive to cell spreading and differentiation following initial cell capture are key to the eventual formation of a functional endothelium. In this study, syndecan-4 and stromal cell-derived factor-1 alpha were used to functionalize an elastomeric biomaterial composed of poly(glycerol sebacate), Silk Fibroin and Type I Collagen, termed PFC, to enhance ECFC-material interaction. Functionalized PFC (fPFC) showed significantly greater ECFCs capture capability under physiological flow. Individual cell spreading area on fPFC (1474 ± 63 μm2 ) was significantly greater than on PFC (1187 ± 54 μm2 ) as early as 2 h, indicating enhanced cell-material interaction. Moreover, fPFC significantly upregulated the expression of endothelial cell specific markers such as platelet endothelial cell adhesion molecule (24-fold) and Von Willebrand Factor (11-fold) compared with tissue culture plastic after 7 days, demonstrating differentiation of ECFCs into endothelial cells. fPFC fabricated as small diameter conduits and tested using a pulsatile blood flow bioreactor were stable and maintained function. The findings suggest that the new surface functionalization strategy proposed here results in an endovascular material with enhanced endothelialization.
Collapse
Affiliation(s)
- Yidi Wu
- Department of Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Engineering and Sciences, Virginia Tech - Wake Forest University School, Winston-Salem, North Carolina, USA
| | - Saami K Yazdani
- Department of Engineering, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Johanna Elin Marie Bolander
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - William D Wagner
- Department of Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Engineering and Sciences, Virginia Tech - Wake Forest University School, Winston-Salem, North Carolina, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| |
Collapse
|
24
|
Zhang F, Tao H, Gluck JM, Wang L, Daneshmand MA, King MW. A textile-reinforced composite vascular graft that modulates macrophage polarization and enhances endothelial cell migration, adhesion and proliferation in vitro. SOFT MATTER 2023; 19:1624-1641. [PMID: 36752696 DOI: 10.1039/d2sm01190e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
At the present time, there is no successful off-the-shelf small-caliber vascular graft (<6 mm) for the repair or bypass of the coronary or carotid arteries. In this study, we engineer a textile-reinforced hydrogel vascular graft. The textile fibers are circularly knitted into a flexible yet robust conduit to serve as the backbone of the composite vascular graft and provide the primary mechanical support. It is embedded in the hydrogel matrix which seals the open structure of the knitted reinforcement and mediates cellular response toward a faster reendothelialization. The mechanical properties of the composite vascular graft, including bursting strength, suture retention strength and radial compliance, significantly surpass the requirement for the vascular graft application and can be adjusted by altering the structure of the textile reinforcement. The addition of hydrogel matrix, on the other hand, improves the survival, adhesion and proliferation of endothelial cells in vitro. The composite vascular graft also enhances macrophage activation and upregulates M1 and M2 related gene expression, which further improves the endothelial cell migration that might favor the reendothelialization of the vascular graft. Taken together, the textile-reinforced hydrogel shows it potential to be a promising scaffold material to fabricate a tissue engineered vascular graft.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of Textiles, North Carolina State University, Raleigh, NC 27606, USA.
| | - Hui Tao
- College of Textiles, Donghua University, Shanghai, 201620, China
| | - Jessica M Gluck
- Wilson College of Textiles, North Carolina State University, Raleigh, NC 27606, USA.
| | - Lu Wang
- College of Textiles, Donghua University, Shanghai, 201620, China
| | - Mani A Daneshmand
- Department of Surgery, School of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Martin W King
- Wilson College of Textiles, North Carolina State University, Raleigh, NC 27606, USA.
- College of Textiles, Donghua University, Shanghai, 201620, China
| |
Collapse
|
25
|
Tan W, Boodagh P, Selvakumar PP, Keyser S. Strategies to counteract adverse remodeling of vascular graft: A 3D view of current graft innovations. Front Bioeng Biotechnol 2023; 10:1097334. [PMID: 36704297 PMCID: PMC9871289 DOI: 10.3389/fbioe.2022.1097334] [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: 11/13/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Vascular grafts are widely used for vascular surgeries, to bypass a diseased artery or function as a vascular access for hemodialysis. Bioengineered or tissue-engineered vascular grafts have long been envisioned to take the place of bioinert synthetic grafts and even vein grafts under certain clinical circumstances. However, host responses to a graft device induce adverse remodeling, to varied degrees depending on the graft property and host's developmental and health conditions. This in turn leads to invention or failure. Herein, we have mapped out the relationship between the design constraints and outcomes for vascular grafts, by analyzing impairment factors involved in the adverse graft remodeling. Strategies to tackle these impairment factors and counteract adverse healing are then summarized by outlining the research landscape of graft innovations in three dimensions-cell technology, scaffold technology and graft translation. Such a comprehensive view of cell and scaffold technological innovations in the translational context may benefit the future advancements in vascular grafts. From this perspective, we conclude the review with recommendations for future design endeavors.
Collapse
Affiliation(s)
- Wei Tan
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States,*Correspondence: Wei Tan,
| | - Parnaz Boodagh
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Sean Keyser
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
26
|
Mazloomnejad R, Babajani A, Kasravi M, Ahmadi A, Shariatzadeh S, Bahrami S, Niknejad H. Angiogenesis and Re-endothelialization in decellularized scaffolds: Recent advances and current challenges in tissue engineering. Front Bioeng Biotechnol 2023; 11:1103727. [PMID: 36873356 PMCID: PMC9978201 DOI: 10.3389/fbioe.2023.1103727] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
Decellularization of tissues and organs has recently become a promising approach in tissue engineering and regenerative medicine to circumvent the challenges of organ donation and complications of transplantations. However, one main obstacle to reaching this goal is acellular vasculature angiogenesis and endothelialization. Achieving an intact and functional vascular structure as a vital pathway for supplying oxygen and nutrients remains the decisive challenge in the decellularization/re-endothelialization procedure. In order to better understand and overcome this issue, complete and appropriate knowledge of endothelialization and its determining variables is required. Decellularization methods and their effectiveness, biological and mechanical characteristics of acellular scaffolds, artificial and biological bioreactors, and their possible applications, extracellular matrix surface modification, and different types of utilized cells are factors affecting endothelialization consequences. This review focuses on the characteristics of endothelialization and how to optimize them, as well as discussing recent developments in the process of re-endothelialization.
Collapse
Affiliation(s)
- Radman Mazloomnejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Kasravi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armin Ahmadi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, United States
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Trujillo-Miranda M, Apsite I, Agudo JAR, Constante G, Ionov L. 4D Biofabrication of Mechanically Stable Tubular Constructs Using Shape Morphing Porous Bilayers for Vascularization Application. Macromol Biosci 2023; 23:e2200320. [PMID: 36165235 DOI: 10.1002/mabi.202200320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/11/2022] [Indexed: 01/19/2023]
Abstract
This study reports the fabrication of highly porous electrospun self-folding bilayers, which fold into tubular structures with excellent mechanical stability, allowing them to be easily manipulated and handled. Two kinds of bilayers based on biocompatible and biodegradable soft (PCL, polycaprolactone) and hard (PHB, poly-hydroxybutyrate) thermoplastic polymers have been fabricated and compared. Multi-scroll structures with tunable diameter are obtained after the shape transformation of the bilayer in aqueous media, where PCL-based bilayer rolled longitudinally and PHB-based one rolled transversely with respect to the fiber direction. A combination of higher elastic modulus and transverse orientation of fibers with respect to rolling direction allowed precise temporal control of shape transformation of PHB-bilayer - stress produced by swollen methacrylated hyaluronic acid (HA-MA) do not relax with time and folding is not affected by the fact that bilayer is fixed in unfolded state in cell culture medium for more than 1 h. This property of PHB-bilayer allowed cell culturing without a negative effect on its shape transformation ability. Moreover, PHB-based tubular structure demonstrated superior mechanical stability compared to PCL-based ones and do not collapse during manipulations that happened to PCL-based one. Additionally, PHB/HA-MA bilayers showed superior biocompatibility, degradability, and long-term stability compared to PCL/HA-MA.
Collapse
Affiliation(s)
- Mairon Trujillo-Miranda
- Faculty of Engineering Sciences and Bavarian Polymer Institute, University of Bayreuth, Ludwig Thoma Str. 36A, 95447, Bayreuth, Germany
| | - Indra Apsite
- Faculty of Engineering Sciences and Bavarian Polymer Institute, University of Bayreuth, Ludwig Thoma Str. 36A, 95447, Bayreuth, Germany
| | | | - Gissela Constante
- Faculty of Engineering Sciences and Bavarian Polymer Institute, University of Bayreuth, Ludwig Thoma Str. 36A, 95447, Bayreuth, Germany
| | - Leonid Ionov
- Faculty of Engineering Sciences and Bavarian Polymer Institute, University of Bayreuth, Ludwig Thoma Str. 36A, 95447, Bayreuth, Germany
| |
Collapse
|
28
|
Liang X, Lei Y, Ding K, Huang X, Zheng C, Wang Y. Poly(2-methoxyethyl acrylate) coated bioprosthetic heart valves by copolymerization with enhanced anticoagulant, anti-inflammatory, and anti-calcification properties. J Mater Chem B 2022; 10:10054-10064. [PMID: 36448545 DOI: 10.1039/d2tb01826h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Commercial glutaraldehyde (Glut) cross-linked bioprosthetic heart valves (BHVs) fabricated from the pericardium have become the most popular choice for treating heart valve diseases. Nevertheless, thrombosis, inflammation and calcification might lead to structural valve degeneration (SVD), which limited the durability of BHVs. Herein, to improve the biocompatibility of BHVs, we fabricated a poly-(2-methoxyethyl acrylate) (PMEA) coated porcine pericardium (PMEA-PP) through grafting PMEA to the porcine pericardium (PP) that was pre-treated with Glut and methacrylated polylysine. PMEA coating mitigated the side effects caused by aldehyde residues. It was shown that the PMEA coating reduced cytotoxicity and inflammation reactions and improved endothelialization potential, and its hydrophilic surface improved the anti-thrombotic properties of PPs. And the PMEA coating significantly reduced the calcification of PPs. This strategy promoted the endothelialization potential and improve the anti-thrombosis and anti-calcification properties of BHVs, and is expected to overcome the defects of commercial BHVs.
Collapse
Affiliation(s)
- Xuyue Liang
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Yang Lei
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Kailei Ding
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Xueyu Huang
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Cheng Zheng
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, No. 29 Wangjiang Road, Chengdu 610064, P. R. China.
| |
Collapse
|
29
|
Nasiri B, Yi T, Wu Y, Smith RJ, Podder AK, Breuer CK, Andreadis ST. Monocyte Recruitment for Vascular Tissue Regeneration. Adv Healthc Mater 2022; 11:e2200890. [PMID: 36112115 PMCID: PMC9671850 DOI: 10.1002/adhm.202200890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/05/2022] [Indexed: 01/28/2023]
Abstract
A strategy to recruit monocytes (MCs) from blood to regenerate vascular tissue from unseeded (cell-free) tissue engineered vascular grafts is presented. When immobilized on the surface of vascular grafts, the fusion protein, H2R5 can capture blood-derived MC under static or flow conditions in a shear stress dependent manner. The bound MC turns into macrophages (Mϕ) expressing both M1 and M2 phenotype specific genes. When H2R5 functionalized acellular-tissue engineered vessels (A-TEVs) are implanted into the mouse aorta, they remain patent and form a continuous endothelium expressing both endothelial cell (EC) and MC specific proteins. Underneath the EC layer, multiple cells layers are formed coexpressing both smooth muscle cell (SMC) and MC specific markers. Lineage tracing analysis using a novel CX3CR1-confetti mouse model demonstrates that fluorescently labeled MC populates the graft lumen by two and four weeks postimplantation, providing direct evidence in support of MC/Mϕ recruitment to the graft lumen. Given their abundance in the blood, circulating MCs may be a great source of cells that contribute directly to the endothelialization and vascular wall formation of acellular vascular grafts under the right chemical and biomechanical cues.
Collapse
Affiliation(s)
- Bita Nasiri
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Tai Yi
- Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Yulun Wu
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Randall J. Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Ashis Kumar Podder
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | | | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY
- Center for Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| |
Collapse
|
30
|
Wang Z, Mithieux SM, Vindin H, Wang Y, Zhang M, Liu L, Zbinden J, Blum KM, Yi T, Matsuzaki Y, Oveissi F, Akdemir R, Lockley KM, Zhang L, Ma K, Guan J, Waterhouse A, Pham NTH, Hawkett BS, Shinoka T, Breuer CK, Weiss AS. Rapid Regeneration of a Neoartery with Elastic Lamellae. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2205614. [PMID: 36120809 DOI: 10.1002/adma.202205614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Native arteries contain a distinctive intima-media composed of organized elastin and an adventitia containing mature collagen fibrils. In contrast, implanted biodegradable small-diameter vascular grafts do not present spatially regenerated, organized elastin. The elastin-containing structures within the intima-media region encompass the elastic lamellae (EL) and internal elastic lamina (IEL) and are crucial for normal arterial function. Here, the development of a novel electrospun small-diameter vascular graft that facilitates de novo formation of a structurally appropriate elastin-containing intima-media region following implantation is described. The graft comprises a non-porous microstructure characterized by tropoelastin fibers that are embedded in a PGS matrix. After implantation in mouse abdominal aorta, the graft develops distinct cell and extracellular matrix profiles that approximate the native adventitia and intima-media by 8 weeks. Within the newly formed intima-media region there are circumferentially aligned smooth muscle cell layers that alternate with multiple EL similar to that found in the arterial wall. By 8 months, the developed adventitia region contains mature collagen fibrils and the neoartery presents a distinct IEL with thickness comparable to that in mouse abdominal aorta. It is proposed that this new class of material can generate the critically required, organized elastin needed for arterial regeneration.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Suzanne M Mithieux
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Howard Vindin
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Yiwei Wang
- Burns Research and Reconstructive Surgery, Anzac Research Institute, Sydney, NSW, 2139, Australia
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Miao Zhang
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Linyang Liu
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Jacob Zbinden
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Kevin M Blum
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Tai Yi
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Yuichi Matsuzaki
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Farshad Oveissi
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Reyda Akdemir
- Department of Chemical Engineering, University Rovira i Virgili, Tarragona, E-43007, Spain
| | - Karen M Lockley
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Lingyue Zhang
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science and Engineering, Beihang University, Beijing, 100191, China
| | - Ke Ma
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science and Engineering, Beihang University, Beijing, 100191, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science and Engineering, Beihang University, Beijing, 100191, China
| | - Anna Waterhouse
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
- School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
- The Heart Research Institute, University of Sydney, Sydney, NSW, 204206, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Nguyen T H Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW, 2006, Australia
| | - Brian S Hawkett
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney, Sydney, NSW, 2006, Australia
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Anthony S Weiss
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
31
|
Exarchos V, Zacharova E, Neuber S, Giampietro C, Motta SE, Hinkov H, Emmert MY, Nazari-Shafti TZ. The path to a hemocompatible cardiovascular implant: Advances and challenges of current endothelialization strategies. Front Cardiovasc Med 2022; 9:971028. [PMID: 36186971 PMCID: PMC9515323 DOI: 10.3389/fcvm.2022.971028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular (CV) implants are still associated with thrombogenicity due to insufficient hemocompatibility. Endothelialization of their luminal surface is a promising strategy to increase their hemocompatibility. In this review, we provide a collection of research studies and review articles aiming to summarize the recent efforts on surface modifications of CV implants, including stents, grafts, valves, and ventricular assist devises. We focus in particular on the implementation of micrometer or nanoscale surface modifications, physical characteristics of known biomaterials (such as wetness and stiffness), and surface morphological features (such as gratings, fibers, pores, and pits). We also review how biomechanical signals originating from the endothelial cell for surface interaction can be directed by topography engineering approaches toward the survival of the endothelium and its long-term adaptation. Finally, we summarize the regulatory and economic challenges that may prevent clinical implementation of endothelialized CV implants.
Collapse
Affiliation(s)
- Vasileios Exarchos
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Ema Zacharova
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Department of Life Sciences, IMC University of Applied Sciences Krems, Krems an der Donau, Austria
| | - Sebastian Neuber
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Costanza Giampietro
- Experimental Continuum Mechanics, Empa Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, Switzerland
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zürich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Hristian Hinkov
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Maximilian Y. Emmert
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Clinic for Cardiovascular Surgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Timo Z. Nazari-Shafti
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin, Germany
- *Correspondence: Timo Z. Nazari-Shafti,
| |
Collapse
|
32
|
Tian Y, Seeto WJ, Páez-Arias MA, Hahn MS, Lipke EA. Endothelial colony forming cell rolling and adhesion supported by peptide-grafted hydrogels. Acta Biomater 2022; 152:74-85. [PMID: 36031035 DOI: 10.1016/j.actbio.2022.08.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 07/28/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023]
Abstract
The aim of this study was to investigate the ability of peptides and peptide combinations to support circulating endothelial colony forming cell (ECFC) rolling and adhesion under shear flow, informing biomaterial design in moving toward rapid cardiovascular device endothelialization. ECFCs have high proliferative capability and can differentiate into endothelial cells, making them a promising cell source for endothelialization. Both single peptides and peptide combinations designed to target integrins α4β1 and α5β1 were coupled to poly(ethylene glycol) hydrogels, and their performance was evaluated by monitoring velocity patterns during the ECFC rolling process, in addition to firm adhesion (capture). Tether percentage and velocity fluctuation, a parameter newly defined here, were found to be valuable in assessing cell rolling velocity patterns and when used in combination were able to predict cell capture. REDV-containing peptides binding integrin α4β1 have been previously shown to reduce ECFC rolling velocity but not to support firm adhesion. This study finds that the performance of REDV-containing peptides in facilitating ECFC dynamic adhesion and capture can be improved by combination with α5β1 integrin-binding peptides, which support ECFC static adhesion. Moreover, when similar in length, the peptide combinations may have synergistic effects in capturing ECFCs. With matching lengths, the peptide combinations including CRRETAWAC(cyclic)+REDV, P_RGDS+KSSP_REDV, and P_RGDS+P_REDV showed high values in both tether percentage and velocity fluctuation and improvement in ECFC capture compared to the single peptides at the shear rate of 20 s-1. These newly identified peptide combinations have the potential to be used as vascular device coatings to recruit ECFCs. STATEMENT OF SIGNIFICANCE: Restoration of functional endothelium following placement of stents and vascular grafts is critical for maintaining long-term patency. Endothelial colony forming cells (ECFCs) circulating in blood flow are a valuable cell source for rapid endothelialization. Here we identify and test novel peptides and peptide combinations that can potentially be used as coatings for vascular devices to support rolling and capture of ECFCs from flow. In addition to the widely used assessment of final ECFC adhesion, we also recorded the rolling process to quantitatively evaluate the interaction between ECFCs and the peptides, obtaining detailed performance of the peptides and gaining insight into effective capture molecule design. Peptide combinations targeting both integrin α4β1 and integrin α5β1 showed the highest percentages of ECFC capture.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Wen J Seeto
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Mayra A Páez-Arias
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Mariah S Hahn
- Biomedical Engineering Department, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA.
| |
Collapse
|
33
|
Inflammation-triggered dual release of nitroxide radical and growth factor from heparin mimicking hydrogel-tissue composite as cardiovascular implants for anti-coagulation, endothelialization, anti-inflammation, and anti-calcification. Biomaterials 2022; 289:121761. [DOI: 10.1016/j.biomaterials.2022.121761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/20/2022]
|
34
|
Xiao W, Chen W, Wang Y, Zhang C, Zhang X, Zhang S, Wu W. Recombinant DTβ4-inspired porous 3D vascular graft enhanced antithrombogenicity and recruited circulating CD93 +/CD34 + cells for endothelialization. SCIENCE ADVANCES 2022; 8:eabn1958. [PMID: 35857526 PMCID: PMC9278867 DOI: 10.1126/sciadv.abn1958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/27/2022] [Indexed: 05/31/2023]
Abstract
Matching material degradation with host remodeling, including endothelialization and muscular remodeling, is important to vascular regeneration. We fabricated 3D PGS-PCL vascular grafts, which presented tunable polymer components, porosity, mechanical strength, and degrading rate. Furthermore, highly porous structures enabled 3D patterning of conjugated heparin-binding peptide, dimeric thymosin β4 (DTβ4), which played key roles in antiplatelets, fibrinogenesis inhibition, and recruiting circulating progenitor cells, thereafter contributed to high patency rate, and unprecedentedly acquired carotid arterial regeneration in rabbit model. Through single-cell RNA sequencing analysis and cell tracing studies, a subset of endothelial progenitor cells, myeloid-derived CD93+/CD34+ cells, was identified as the main contributor to final endothelium regeneration. To conclude, DTβ4-inspired porous 3DVGs present adjustable physical properties, superior anticoagulating, and re-endothelializing potentials, which leads to the regeneration of small-caliber artery, thus offering a promising tool for vessel replacement in clinical applications.
Collapse
Affiliation(s)
- Weiwei Xiao
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Wanli Chen
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Yinggang Wang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Xinchi Zhang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Siqian Zhang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Wei Wu
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
35
|
Yi B, Zhou B, Song Z, Yu L, Wang W, Liu W. Step-wise CAG@PLys@PDA-Cu2+ modification on micropatterned nanofibers for programmed endothelial healing. Bioact Mater 2022; 25:657-676. [PMID: 37056258 PMCID: PMC10086768 DOI: 10.1016/j.bioactmat.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022] Open
Abstract
Native-like endothelium regeneration is a prerequisite for material-guided small-diameter vascular regeneration. In this study, a novel strategy is proposed to achieve phase-adjusted endothelial healing by step-wise modification of parallel-microgroove-patterned (i.e., micropatterned) nanofibers with polydopamine-copper ion (PDA-Cu2+) complexes, polylysine (PLys) molecules, and Cys-Ala-Gly (CAG) peptides (CAG@PLys@PDA-Cu2+). Using electrospun poly(l-lactide-co-caprolactone) random nanofibers as the demonstrating biomaterial, step-wise modification of CAG@PLys@PDA-Cu2+ significantly enhanced substrate wettability and protein adsorption, exhibited an excellent antithrombotic surface and outstanding phase-adjusted capacity of endothelium regeneration involving cell adhesion, endothelial monolayer formation, and the regenerated endothelium maturation. Upon in vivo implantation for segmental replacement of rabbit carotid arteries, CAG@PLys@PDA-Cu2+ modified grafts (2 mm inner diameter) with micropatterns on inner surface effectively accelerated native-like endothelium regeneration within 1 week, with less platelet aggregates and inflammatory response compared to those on non-modified grafts. Prolonged observations at 6- and 12-weeks post-implantation demonstrated a positive vascular remodeling with almost fully covered endothelium and mature smooth muscle layer in the modified vascular grafts, accompanied with well-organized extracellular matrix. By contrast, non-modified vascular grafts induced a disorganized tissue formation with a high risk of thrombogenesis. In summary, step-wise modification of CAG@PLys@PDA-Cu2+ on micropatterned nanofibers can significantly promote endothelial healing without inflicting thrombosis, thus confirming a novel strategy for developing functional vascular grafts or other blood-contacting materials/devices.
Collapse
|
36
|
Zhang Y, Zhang X, Meng Y, Xu X, Zuo D. The role of glycolysis and lactate in the induction of tumor-associated macrophages immunosuppressive phenotype. Int Immunopharmacol 2022; 110:108994. [PMID: 35777265 DOI: 10.1016/j.intimp.2022.108994] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/30/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022]
Abstract
Growing evidence highlights that glycolysis and tumor-derived lactate could skew tumor-associated macrophages (TAMs) toward an immunosuppressive phenotype. However, the updated research has not been systematically summarized yet. TAMs are educated by the tumor microenvironment (TME) and exert immunosuppressive functions and tumorigenic effects via multiple biological processes. It is well known that lactate generated by aerobic glycolysis is significantly accumulated in TME and promotes tumor progression in solid tumors. Moreover, some recent research demonstrated that glycolysis is activated in TAMs to support M2-like polarization, which is absolutely in contrast with the metabolic profile of M2 macrophages in inflammation. Notably, lactate produced by high levels of glycolysis is not only a metabolic by-product but also an oncometabolite. TAMs could access the biological information delivered by lactate and further enhance protumor functions such as immunosuppression and angiogenesis. Here, we outline the connection between glycolysis and TAM phenotype to elucidate the metabolic characteristics of TAMs. Further, insights into the specific molecular mechanisms of lactate-induced TAM polarization and potential therapeutic targets are summarized. We sought to discuss the reciprocal interaction between tumor cells and TAMs mediated by lactate, which will lay a foundation for the research aiming to elucidate the complex functions of TAMs.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yuting Meng
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiaobo Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| |
Collapse
|
37
|
Kraus X, van de Flierdt E, Renzelmann J, Thoms S, Witt M, Scheper T, Blume C. Peripheral blood derived endothelial colony forming cells as suitable cell source for pre-endothelialization of arterial vascular grafts under dynamic flow conditions. Microvasc Res 2022; 143:104402. [PMID: 35753506 DOI: 10.1016/j.mvr.2022.104402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022]
Abstract
In regenerative medicine, autologous peripheral blood derived endothelial colony forming cells (PB-derived ECFC) represent a promising source of endothelial cells (EC) for pre-endothelialization of arterial tissue engineered vascular grafts (TEVG) since they are readily attainable, can easily be isolated and possess a high proliferation potential. The aim of this study was to compare the phenotype of PB-derived ECFC with arterial and venous model cells such as human aortic endothelial cells (HAEC) and human umbilical vein endothelial cells (HUVEC) under dynamic cell culture conditions to find a suitable cell source of EC for pre-endothelialization. In this study PB-derived ECFC were cultivated over 24 h under a high pulsatile shear stress (20 dyn/cm2, 1 Hz) and subsequently analyzed. ECFC oriented and elongated in the direction of flow and expressed similar anti-thrombotic and endothelial differentiation markers compared to HAEC. There were significant differences observable in gene expression levels of CD31, CD34 and NOTCH4 between ECFC and HUVEC. These results therefore suggest an arterial phenotype for PB-derived ECFC both under static and flow conditions, and this was supported by NOTCH4 protein expression profiles. ECFC also significantly up-regulated gene expression levels of anti-thrombotic genes such as krueppel-like factor 2, endothelial nitric oxide synthase 3 and thrombomodulin under shear stress cultivation as compared to static conditions. Dynamically cultured PB-derived ECFC therefore may be a promising cell source for pre-endothelialization of arterial TEVGs.
Collapse
Affiliation(s)
- Xenia Kraus
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany.
| | - Edda van de Flierdt
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Jannis Renzelmann
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Stefanie Thoms
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Martin Witt
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Thomas Scheper
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Cornelia Blume
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| |
Collapse
|
38
|
Zhang F, King MW. Immunomodulation Strategies for the Successful Regeneration of a Tissue-Engineered Vascular Graft. Adv Healthc Mater 2022; 11:e2200045. [PMID: 35286778 PMCID: PMC11468936 DOI: 10.1002/adhm.202200045] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/18/2022] [Indexed: 01/02/2023]
Abstract
Cardiovascular disease leads to the highest morbidity worldwide. There is an urgent need to solve the lack of a viable arterial graft for patients requiring coronary artery bypass surgery. The current gold standard is to use the patient's own blood vessel, such as a saphenous vein graft. However, some patients do not have appropriate vessels to use because of systemic disease or secondary surgery. On the other hand, there is no commercially available synthetic vascular graft available on the market for small diameter (<6 mm) blood vessels like coronary, carotid, and peripheral popliteal arteries. Tissue-engineered vascular grafts (TEVGs) are studied in recent decades as a promising alternative to synthetic arterial prostheses. Yet only a few studies have proceeded to a clinical trial. Recent studies have uncovered that the host immune response can be directed toward increasing the success of a TEVG by shedding light on ways to modulate the macrophage response and improve the tissue regeneration outcome. In this review, the basic concepts of vascular tissue engineering and immunoengineering are considered. The state-of-art of TEVGs is summarized and the role of macrophages in TEVG regeneration is analyzed. Current immunomodulatory strategies based on biomaterials are also discussed.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of TextilesNorth Carolina State UniversityRaleighNC27606USA
| | - Martin W. King
- Wilson College of TextilesNorth Carolina State UniversityRaleighNC27606USA
| |
Collapse
|
39
|
Chen W, Xiao W, Liu X, Yuan P, Zhang S, Wang Y, Wu W. Pharmacological manipulation of macrophage autophagy effectively rejuvenates the regenerative potential of biodegrading vascular graft in aging body. Bioact Mater 2022; 11:283-299. [PMID: 34977432 PMCID: PMC8668428 DOI: 10.1016/j.bioactmat.2021.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/28/2021] [Accepted: 09/22/2021] [Indexed: 12/25/2022] Open
Abstract
Declined regenerative potential and aggravated inflammation upon aging create an inappropriate environment for arterial regeneration. Macrophages are one of vital effector cells in the immune microenvironment, especially during biomaterials mediated repairing process. Here, we revealed that the macrophage autophagy decreased with aging, which led to aggravated inflammation, thereby causing poor vascular remodeling of artificial grafts in aging body. Through loading the autophagy-targeted drugs, rapamycin and 3-MA (3-methyladenine), in PCL (polycaprolactone) sheath of the PGS (poly glycerol sebacate) - PCL vascular graft, the essential role of macrophage autophagy was confirmed in regulating macrophage polarization and biomaterial degradation. Moreover, the utilization of rapamycin promoted anti-inflammatory polarization of macrophage by activating autophagy, which further promoted myogenic differentiation of vascular progenitor cells and accelerated endothelialization. Our study elucidated the contribution of pharmacological manipulation of macrophage autophagy in promoting regeneration of small caliber artery, which may pave a new avenue for clinical translation of vascular grafts in aging body.
Collapse
Affiliation(s)
- Wanli Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 399 Middle Yanchang Road, Shanghai 200072, China
| | - Weiwei Xiao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xuzheng Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pingping Yuan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Siqian Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yinggang Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
40
|
Cheng S, Liu X, Qian Y, Maitusong M, Yu K, Cao N, Fang J, Liu F, Chen J, Xu D, Zhu G, Ren T, Wang J. Double-Network Hydrogel Armored Decellularized Porcine Pericardium as Durable Bioprosthetic Heart Valves. Adv Healthc Mater 2022; 11:e2102059. [PMID: 34969157 DOI: 10.1002/adhm.202102059] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/02/2021] [Indexed: 12/20/2022]
Abstract
Heart valves have extraordinary fatigue resistance which beat ≈3 billion times in a lifetime. Bioprosthetic heart valves (BHVs) made from fixed heteroplasm that are incrementally used in heart valve replacement fail to sustain the expected durability due to thrombosis, poor endothelialization, inflammation, calcification, and especially mechanical damage induced biocompatibility change. No effective strategy has been reported to conserve the biological properties of BHV after long-term fatigue test. Here, a double-network tough hydrogel is introduced, which interpenetrate and anchor into the matrix of decellularized porcine pericardium (dCell-PP) to form robust and stable conformal coatings and reduce immunogenicity. The ionic crosslinked hyaluronic acid (HA) network mimics the glycocalyx on endothelium which improves antithrombosis and accelerates endothelialization; the chemical crosslinked hydrophilic polyacrylamide (PAAm) network further enhances antifouling properties and strengthens the shielding hydrogels and their interaction with dCell-PP. In vitro and rabbit ex vivo shunt assay demonstrate great hemocompatibility of polyacrylamide/HA hydrogel hybrid PP (P/H-PP). Cell experiments and rat subcutaneous implantation confirm satisfactory endothelialization, biocompatibility, and anticalcification properties. For hydrodynamic experiment, P/H-PP gains full mark at different flow conditions and sustains excellent biomechanical and biological properties after 200 000 000 cycles. P/H double-network hydrogel armoring dCell-PP is a promising progress to extend BHV durability for clinical implantation therapy.
Collapse
Affiliation(s)
- Si Cheng
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Xianbao Liu
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Yi Qian
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Miribani Maitusong
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Kaixiang Yu
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Naifang Cao
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Juan Fang
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Feng Liu
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Jinyong Chen
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Dilin Xu
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Gangjie Zhu
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Tanchen Ren
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| | - Jian'an Wang
- Department of Cardiology of The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Cardiovascular Key Laboratory of Zhejiang Province Hangzhou 310009 P. R. China
| |
Collapse
|
41
|
Shah P, Chandra S. Review on emergence of nanomaterial coatings in bio-engineered cardiovascular stents. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Zakharova I, Saaya S, Shevchenko A, Stupnikova A, Zhiven' M, Laktionov P, Stepanova A, Romashchenko A, Yanshole L, Chernonosov A, Volkov A, Kizilova E, Zavjalov E, Chernyavsky A, Romanov A, Karpenko A, Zakian S. Mitomycin-Treated Endothelial and Smooth Muscle Cells Suitable for Safe Tissue Engineering Approaches. Front Bioeng Biotechnol 2022; 10:772981. [PMID: 35360387 PMCID: PMC8963790 DOI: 10.3389/fbioe.2022.772981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
In our previous study, we showed that discarded cardiac tissue from the right atrial appendage and right ventricular myocardium is an available source of functional endothelial and smooth muscle cells for regenerative medicine and tissue engineering. In the study, we aimed to find out what benefits are given by vascular cells from cardiac explants used for seeding on vascular patches engrafted to repair vascular defects in vivo. Additionally, to make the application of these cells safer in regenerative medicine we tested an in vitro approach that arrested mitotic division to avoid the potential tumorigenic effect of dividing cells. A tissue-engineered construction in the form of a patch based on a polycaprolactone-gelatin scaffold and seeded with endothelial and smooth muscle cells was implanted into the abdominal aorta of immunodeficient SCID mice. Aortic patency was assessed using ultrasound, MRI, immunohistochemical and histological staining. Endothelial and smooth muscle cells were treated with mitomycin C at a therapeutic concentration of 10 μg/ml for 2 h with subsequent analysis of cell proliferation and function. The absence of the tumorigenic effect of mitomycin C-treated cells, as well as their angiogenic potential, was examined by injecting them into immunodeficient mice. Cell-containing patches engrafted in the abdominal aorta of immunodeficient mice form the vessel wall loaded with the appropriate cells and extracellular matrix, and do not interfere with normal patency. Endothelial and smooth muscle cells treated with mitomycin C show no tumorigenic effect in the SCID immunodeficient mouse model. During in vitro experiments, we have shown that treatment with mitomycin C does not lead to a decrease in cell viability. Despite the absence of proliferation, mitomycin C-treated vascular cells retain specific cell markers, produce specific extracellular matrix, and demonstrate the ability to stimulate angiogenesis in vivo. We pioneered an approach to arresting cell division with mitomycin C in endothelial and smooth muscle cells from cardiac explant, which prevents the risk of malignancy from dividing cells in vascular surgery. We believe that this approach to the fabrication of tissue-engineered constructs based on mitotically inactivated cells from waste postoperative material may be valuable to bring closer the development of safe cell products for regenerative medicine.
Collapse
Affiliation(s)
- Irina Zakharova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- *Correspondence: Irina Zakharova,
| | - Shoraan Saaya
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Alexander Shevchenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Stupnikova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Deparment of Natural Science, Novosibirsk State University, Novosibirsk, Russia
| | - Maria Zhiven'
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Pavel Laktionov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Stepanova
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Romashchenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Lyudmila Yanshole
- International Tomography Center,The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Chernonosov
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Volkov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Elena Kizilova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Deparment of Natural Science, Novosibirsk State University, Novosibirsk, Russia
| | - Evgenii Zavjalov
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Chernyavsky
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Alexander Romanov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Andrey Karpenko
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Suren Zakian
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
43
|
Failure Analysis of TEVG’s II: Late Failure and Entering the Regeneration Pathway. Cells 2022; 11:cells11060939. [PMID: 35326390 PMCID: PMC8946846 DOI: 10.3390/cells11060939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) are a promising alternative to treat vascular disease under complex hemodynamic conditions. However, despite efforts from the tissue engineering and regenerative medicine fields, the interactions between the material and the biological and hemodynamic environment are still to be understood, and optimization of the rational design of vascular grafts is an open challenge. This is of special importance as TEVGs not only have to overcome the surgical requirements upon implantation, they also need to withhold the inflammatory response and sustain remodeling of the tissue. This work aims to analyze and evaluate the bio-molecular interactions and hemodynamic phenomena between blood components, cells and materials that have been reported to be related to the failure of the TEVGs during the regeneration process once the initial stages of preimplantation have been resolved, in order to tailor and refine the needed criteria for the optimal design of TEVGs.
Collapse
|
44
|
Lau S, Gossen M, Lendlein A, Jung F. Differential sensitivity of assays for determining vein endothelial cell senescence. Clin Hemorheol Microcirc 2022; 81:191-203. [PMID: 35275526 DOI: 10.3233/ch-211294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In vivo endothelialization of polymer-based cardiovascular implant materials is a promising strategy to reduce the risk of platelet adherence and the subsequent thrombus formation and implant failure. However, endothelial cells from elderly patients are likely to exhibit a senescent phenotype that may counteract endothelialization. The senescence status of cells should therefore be investigated prior to implantation of devices designed to be integrated in the blood vessel wall. Here, human umbilical vein endothelial cells (HUVEC) were cultivated up to passage (P) 4, 10 and 26/27 to determine the population doubling time and the senescence status by four different methods. Determination of the senescence-associated β-galactosidase activity (SA-β-Gal) was carried out by colorimetric staining and microscopy (i), as well as by photometric quantification (ii), and the expression of senescence-associated nuclear proteins p16 and p21 as well as the proliferation marker Ki67 was assessed by immunostaining (iii), and by flow cytometry (iv). The population doubling time of P27-cells was remarkably greater (103±65 h) compared to P4-cells (24±3 h) and P10-cell (37±15 h). Among the four different methods tested, the photometric SA-β-Gal activity assay and the flow cytometric determination of p16 and Ki67 were most effective in discriminating P27-cells from P4- and P10-cells. These methods combined with functional endothelial cell analyses might aid predictions on the performance of implant endothelialization in vivo.
Collapse
Affiliation(s)
- S Lau
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - M Gossen
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - A Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - F Jung
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| |
Collapse
|
45
|
Generating Monocyte-Derived Endothelial-like Cells for Vascular Regeneration. Methods Mol Biol 2022; 2375:13-19. [PMID: 34591295 PMCID: PMC10013694 DOI: 10.1007/978-1-0716-1708-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A major limitation in engineering vascular grafts is the lack of proper endothelium to prevent thrombosis and subsequent graft failure. Obtaining endothelial cells from patients' vasculature is intrusive and requires extensive culture time. Here we present an alternative strategy wherein abundant and easily accessible monocytes from peripheral blood are cultured and differentiated towards an endothelial-like state capable of preventing thrombosis through production of nitric oxide and formation of endothelial adherens junctions. Considering the plethora of monocytes present within peripheral blood, this method provides a robust alternative to generating endothelial cells required for vascular graft production.
Collapse
|
46
|
Wei Y, Wang F, Guo Z, Zhao Q. Tissue-engineered vascular grafts and regeneration mechanisms. J Mol Cell Cardiol 2021; 165:40-53. [PMID: 34971664 DOI: 10.1016/j.yjmcc.2021.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are life-threatening diseases with high morbidity and mortality worldwide. Vascular bypass surgery is still the ultimate strategy for CVD treatment. Autografts are the gold standard for graft transplantation, but insufficient sources limit their widespread application. Therefore, alternative tissue engineered vascular grafts (TEVGs) are urgently needed. In this review, we summarize the major strategies for the preparation of vascular grafts, as well as the factors affecting their patency and tissue regeneration. Finally, the underlying mechanisms of vascular regeneration that are mediated by host cells are discussed.
Collapse
Affiliation(s)
- Yongzhen Wei
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China; State key Laboratory of Medicinal Chemical Biology & Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Fei Wang
- State key Laboratory of Medicinal Chemical Biology & Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Zhikun Guo
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China
| | - Qiang Zhao
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China; State key Laboratory of Medicinal Chemical Biology & Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
47
|
Immuno-regenerative biomaterials for in situ cardiovascular tissue engineering - Do patient characteristics warrant precision engineering? Adv Drug Deliv Rev 2021; 178:113960. [PMID: 34481036 DOI: 10.1016/j.addr.2021.113960] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023]
Abstract
In situ tissue engineering using bioresorbable material implants - or scaffolds - that harness the patient's immune response while guiding neotissue formation at the site of implantation is emerging as a novel therapy to regenerate human tissues. For the cardiovascular system, the use of such implants, like blood vessels and heart valves, is gradually entering the stage of clinical translation. This opens up the question if and to what extent patient characteristics influence tissue outcomes, necessitating the precision engineering of scaffolds to guide patient-specific neo-tissue formation. Because of the current scarcity of human in vivo data, herein we review and evaluate in vitro and preclinical investigations to predict the potential role of patient-specific parameters like sex, age, ethnicity, hemodynamics, and a multifactorial disease profile, with special emphasis on their contribution to the inflammation-driven processes of in situ tissue engineering. We conclude that patient-specific conditions have a strong impact on key aspects of in situ cardiovascular tissue engineering, including inflammation, hemodynamic conditions, scaffold resorption, and tissue remodeling capacity, suggesting that a tailored approach may be required to engineer immuno-regenerative biomaterials for safe and predictive clinical applicability.
Collapse
|
48
|
Charreau B. Cellular and Molecular Crosstalk of Graft Endothelial Cells During AMR: Effector Functions and Mechanisms. Transplantation 2021; 105:e156-e167. [PMID: 33724240 DOI: 10.1097/tp.0000000000003741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Graft endothelial cell (EC) injury is central to the pathogenesis of antibody-mediated rejection (AMR). The ability of donor-specific antibodies (DSA) to bind C1q and activate the classical complement pathway is an efficient predictor of graft rejection highlighting complement-dependent cytotoxicity as a key process operating during AMR. In the past 5 y, clinical studies further established the cellular and molecular signatures of AMR revealing the key contribution of other, IgG-dependent and -independent, effector mechanisms mediated by infiltrating NK cells and macrophages. Beyond binding to alloantigens, DSA IgG can activate NK cells and mediate antibody-dependent cell cytotoxicity through interacting with Fcγ receptors (FcγRs) such as FcγRIIIa (CD16a). FcRn, a nonconventional FcγR that allows IgG recycling, is highly expressed on ECs and may contribute to the long-term persistence of DSA in blood. Activation of NK cells and macrophages results in the production of proinflammatory cytokines such as TNF and IFNγ that induce transient and reversible changes in the EC phenotype and functions promoting coagulation, inflammation, vascular permeability, leukocyte trafficking. MHC class I mismatch between transplant donor and recipient can create a situation of "missing self" allowing NK cells to kill graft ECs. Depending on the microenvironment, cellular proximity with ECs may participate in macrophage polarization toward an M1 proinflammatory or an M2 phenotype favoring inflammation or vascular repair. Monocytes/macrophages participate in the loss of endothelial specificity in the process of endothelial-to-mesenchymal transition involved in renal and cardiac fibrosis and AMR and may differentiate into ECs enabling vessel and graft (re)-endothelialization.
Collapse
Affiliation(s)
- Béatrice Charreau
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et en Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
49
|
Gupta P, Chaudhuri GR, Janani G, Agarwala M, Ghosh D, Nandi SK, Mandal BB. Functionalized Silk Vascular Grafts with Decellularized Human Wharton's Jelly Improves Remodeling via Immunomodulation in Rabbit Jugular Vein. Adv Healthc Mater 2021; 10:e2100750. [PMID: 34378360 DOI: 10.1002/adhm.202100750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/12/2021] [Indexed: 12/11/2022]
Abstract
Cell-free polymeric tissue-engineered vascular grafts (TEVGs) have shown great promise towards clinical translation; however, their limited bioactivity and remodeling ability challenge this cause. Here, a novel cell-free bioresorbable small diameter silk TEVG system functionalized with decellularized human Wharton's jelly (dWJ) matrix is developed and successfully implanted as interposition grafts into rabbit jugular vein. Implanted TEVGs remain patent for two months and integrate with host tissue, demonstrating neo-tissue formation and constructive remodeling. Mechanistic analysis reveals that dWJ matrix is a reservoir of various immunomodulatory cytokines (Interleukin-8, 6, 10, 4 and tumor necrosis factor alpha (TNF-α)), which aids in upregulating M2 macrophage-associated genes facilitating pro-remodeling behavior. Besides, dWJ treatment to human endothelial cells upregulates the expression of functional genes (cluster of differentiation 31 (CD31), endothelial nitric oxide synthase (eNOS), and vascular endothelial (VE)-cadherin), enables faster cell migration, and elevates nitric oxide (NO) production leading to the in situ development of endothelium. The dWJ functionalized silk TEVGs support increased host cell recruitment than control, including macrophages and vascular cells. It endows superior graft remodeling in terms of a dense medial layer comprising smooth muscle cells and elevates the production of extracellular matrix proteins (collagen and elastin). Altogether, these findings suggest that dWJ functionalization imitates the usefulness of cell seeding and enables graft remodeling.
Collapse
Affiliation(s)
- Prerak Gupta
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Gaurab Ranjan Chaudhuri
- Department of Plastic Surgery R. G. Kar Medical College and Hospital Kolkata West Bengal 700004 India
| | - G. Janani
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Manoj Agarwala
- Department of ENT and Faciomaxillary Surgery GNRC Institute of Medical Sciences Guwahati Assam 781030 India
| | - Debaki Ghosh
- Department of Veterinary Surgery and Radiology West Bengal University of Animal and Fishery Sciences Kolkata West Bengal 700037 India
| | - Samit K. Nandi
- Department of Veterinary Surgery and Radiology West Bengal University of Animal and Fishery Sciences Kolkata West Bengal 700037 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
50
|
Alvino VV, Thomas AC, Ghorbel MT, Rapetto F, Narayan SA, Kilcooley M, Iacobazzi D, Carrabba M, Fagnano M, Cathery W, Avolio E, Caputo M, Madeddu P. Reconstruction of the Swine Pulmonary Artery Using a Graft Engineered With Syngeneic Cardiac Pericytes. Front Bioeng Biotechnol 2021; 9:715717. [PMID: 34568300 PMCID: PMC8459923 DOI: 10.3389/fbioe.2021.715717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
The neonatal heart represents an attractive source of regenerative cells. Here, we report the results of a randomized, controlled, investigator-blinded preclinical study, which assessed the safety and effectiveness of a matrix graft cellularized with cardiac pericytes (CPs) in a piglet model of pulmonary artery (PA) reconstruction. Within each of five trios formed by 4-week-old female littermate piglets, one element (the donor) was sacrificed to provide a source of CPs, while the other two elements (the graft recipients) were allowed to reach the age of 10 weeks. During this time interval, culture-expanded donor CPs were seeded onto swine small intestinal submucosa (SIS) grafts, which were then shaped into conduits and conditioned in a flow bioreactor. Control unseeded SIS conduits were subjected to the same procedure. Then, recipient piglets were randomized to surgical reconstruction of the left PA (LPA) with unseeded or CP-seeded SIS conduits. Doppler echocardiography and cardiac magnetic resonance imaging (CMRI) were performed at baseline and 4-months post-implantation. Vascular explants were examined using histology and immunohistochemistry. All animals completed the scheduled follow-up. No group difference was observed in baseline imaging data. The final Doppler assessment showed that the LPA’s blood flow velocity was similar in the treatment groups. CMRI revealed a mismatch in the average growth of the grafted LPA and contralateral branch in both treatment groups. Histology of explanted arteries demonstrated that the CP-seeded grafts had a thicker luminal cell layer, more intraparietal arterioles, and a higher expression of endothelial nitric oxide synthase (eNOS) compared with unseeded grafts. Moreover, the LPA stump adjacent to the seeded graft contained more elastin and less collagen than the unseeded control. Syngeneic CP engineering did not accomplish the primary goal of supporting the graft’s growth but was able to improve secondary outcomes, such as the luminal cellularization and intraparietal vascularization of the graft, and elastic remodeling of the recipient artery. The beneficial properties of neonatal CPs may be considered in future bioengineering applications aiming to reproduce the cellular composition of native arteries.
Collapse
Affiliation(s)
- Valeria Vincenza Alvino
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Anita C Thomas
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Mohamed T Ghorbel
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Filippo Rapetto
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Srinivas A Narayan
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Michael Kilcooley
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Dominga Iacobazzi
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Michele Carrabba
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Marco Fagnano
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - William Cathery
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|