1
|
Velez FF, Rajani RR, Malone DC, Sun LA, Bloudek L, Carter K, Panaccio M, Niklason LE. Budget impact model of acellular tissue engineered vessel for the repair of extremity arterial trauma when autologous vein is not feasible. J Med Econ 2025; 28:323-334. [PMID: 39964834 DOI: 10.1080/13696998.2025.2469460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/20/2025]
Abstract
AIMS To predict the budget impact of Symvess (Symvess is a trademark of Humacyte Global, Inc.) (acellular tissue engineered vessel-tyod [ATEV]) for extremity arterial trauma repair when autologous vein repair is not feasible. MATERIALS AND METHODS The 3-year budget impact of adding ATEV as a repair option alongside autologous vein, prosthetic graft, and "non-autologous other" grafts was evaluated from the perspectives of a Level I trauma center and third-party commercial payers. Conduit-specific complication rates were obtained from two clinical studies for ATEV and from the published literature and analysis of the PROOVIT registry for other conduits. Costs were compared pre- and post-ATEV availability. Conduit-related costs and complications included conduit infections, amputations, vein harvest site infection, surgical re-interventions, rehabilitation after amputation, and 12-month post-discharge costs. Impact on operating room (OR) time and readmissions was evaluated. A sensitivity analysis was conducted to evaluate parameter uncertainty. RESULTS With introduction of ATEV, there was a 29.8% reduction in amputations and a 29.5% reduction in graft infections over 3 years. From a Level I trauma center perspective, seven patients were expected to receive an ATEV over 3 years, with cumulative cost savings of $80,650 (2.3% decrease). OR time would decrease by 8.6 h, and readmission-related costs would be reduced by 16.7% with ATEV availability. From the third-party commercial payer perspective, 35 patients were expected to receive ATEV, with a budget impact showing a savings of -$0.08 per member per month after 3 years. For trauma centers, sensitivity analysis showed that cost drivers were amputation risk associated with "non-autologous other" graft types and market share of autologous vein (short ischemia time). LIMITATIONS Uncertainty surrounding model parameters. CONCLUSIONS ATEV was projected to be cost-saving over 3 years for both trauma centers and third-party payers due to reductions in the costs related to amputations and conduit infections.
Collapse
Affiliation(s)
| | - Ravi R Rajani
- Department of Surgery, Emory University, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
2
|
Weekes A, Davern JW, Pinto N, Jenkins J, Li Z, Meinert C, Klein TJ. Enhancing compliance and extracellular matrix properties of tissue-engineered vascular grafts through pulsatile bioreactor culture. BIOMATERIALS ADVANCES 2025; 175:214346. [PMID: 40378643 DOI: 10.1016/j.bioadv.2025.214346] [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: 09/19/2024] [Revised: 02/06/2025] [Accepted: 05/11/2025] [Indexed: 05/19/2025]
Abstract
Biofabrication techniques represent a promising avenue for the production of small diameter vascular grafts. However, while current tissue-engineered vascular grafts (TEVGs) fulfil certain functional requirements of native blood vessels, most exhibit very poor mechanical compliance, directly reducing patency in vivo. Here, highly compliant TEVGs were cultured in a dynamic pulsatile bioreactor which ensured enhanced compliance, using biomimetic melt electrowritten (MEW) tubular scaffolds as substrates for tissue growth. Through 6-week in vitro culture, we investigated differences in extracellular matrix (ECM) production and mechanical performance of TEVGs cultured with placental mesenchymal stem cells (MSCs) and smooth muscle cells (SMCs) in static and dynamic conditions. Pulsatile stimulation successfully maintained the high compliance (12.4 ± 0.8 % per 100 mmHg) of our biomimetic scaffolds, substantially greater than existing small diameter grafts. Dynamic TEVGs demonstrated physiologically relevant burst pressure (1125 ± 212 mmHg) and suture pull-out force (3.0 ± 0.4 N), while also accumulating greater ECM components than static TEVGs. To assess off-the-shelf suitability, grafts were decellularized and lyophilised to produce d-TEVGs, which exhibited negligible loss of mechanics or ECM integrity. Finally, rehydrated d-TEVGs were seeded with endothelial cells in vitro, with an intimal endothelial lining forming after 7 days. These findings demonstrate the production of TEVGs with specifically engineered mechanical compliance which has been maintained by dynamic in vitro culture, supporting continued work toward biofabrication of the next generation of vascular grafts.
Collapse
Affiliation(s)
- Angus Weekes
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia; Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia.
| | - Jordan W Davern
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| | - Nigel Pinto
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia; Department of Vascular Surgery, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia.
| | - Jason Jenkins
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia; Department of Vascular Surgery, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia.
| | - Zhiyong Li
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| | - Christoph Meinert
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia; Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia.
| | - Travis J Klein
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
3
|
Al Fahad MA, Lee HY, Park M, Lee BT. A cardiac extracellular matrix-based bilayer vascular graft with controlled microstructures for the reconstruction of small-diameter blood vessels. Biomaterials 2025; 320:123264. [PMID: 40121829 DOI: 10.1016/j.biomaterials.2025.123264] [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: 12/09/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Despite recent progress, challenges with small-diameter vascular grafts, including mechanical strength, intimal hyperplasia, thrombosis, and poor endothelialization, remain unresolved. The present study reports a novel bilayer vascular graft designed to mimic the anatomical features of small-diameter blood vessels. The electrospun graft consists of a dense micro/nanofibrous inner layer of cardiac extracellular matrix (cECM), polycaprolactone (PCL) loaded with heparin (P-cECM-H), and a super porous and micro-fibrous PCL outer layer. Liquid chromatography-mass spectrometry (LC-MS/MS) proteome analysis of the cECM revealed that it is enriched with several bioactive proteins related to angiogenesis, wound regeneration, cell migration, etc. The porosities of the two layers are tailored according to endothelial and smooth muscle cell biology. The graft exhibited excellent mechanical properties, and the heparinized P-cECM inner layer improved hemocompatibility and anticoagulation efficacy. A significant increase in endothelial cell proliferation was noted in the P-cECM-H group after 7 days compared with the control group (p < 0.05). The bilayer graft maintained 100 % patency after 10 weeks of rat abdominal aorta implantation. Histological evaluation revealed smooth muscle cell infiltration inside the highly porous outer layer and neointima regeneration in the inner layer with a complete endothelial lining. RNA sequencing (RNA-Seq) analysis further confirmed smooth muscle formation and endothelial layer formation. The gene expression data also suggested that the hypoxia-inducible factor-1 (HIF-) and vascular endothelial growth factor (VEGF) signaling pathways are involved in endothelial layer remodeling. These promising results indicate that cECM could be a key material for vascular tissue regeneration.
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
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, 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
|
4
|
Henry Dusim GA, Muhamad F, Lai KW. Enhancing Calcium Phosphate Cements: A review of Bacterial Cellulose (BC) and other Biopolymer Reinforcements for Biomedical Applications. BIOMATERIALS ADVANCES 2025; 172:214245. [PMID: 40054229 DOI: 10.1016/j.bioadv.2025.214245] [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: 12/20/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Calcium phosphate cements (CPCs) are renowned for their biocompatibility and osteoconductivity, making them ideal for bone tissue engineering. However, their brittleness and low tensile strength limit their use in load-bearing applications. Bacterial cellulose (BC) has emerged as a promising reinforcement material due to its high tensile strength, biocompatibility, and biodegradability. The incorporation of 2 wt% BC into CPCs increased compressive strength from 5 MPa to 12 MPa, representing a 2.4-fold enhancement, while also improving toughness and promoting cellular interactions through its nanofibrillar structure. Additionally, hybrid composites combining BC with collagen, chitosan, or polycaprolactone (PCL) exhibit synergistic effects, further enhancing mechanical properties and biodegradability. These advancements highlight the potential of BC-reinforced CPCs for clinical applications in bone repair and regeneration. Despite these improvements, limited research addresses tensile and flexural properties, which are critical for load-bearing applications, as well as the effects of BC on injectability and setting time for minimally invasive procedures. Emerging innovations, such as electroactive BC-reinforced CPCs for stimulating bone healing, hold significant potential but remain underexplored. Future research should focus on optimising mechanical properties, validating clinical performance, and developing hybrid formulations to expand their use in load-bearing bone repairs.
Collapse
Affiliation(s)
- Grace Anabela Henry Dusim
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Farina Muhamad
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Khin Wee Lai
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
5
|
Wu Y, Mohamed MA, Yi T, Das A, Rumsey CL, Trebbin M, Breuer CK, Andreadis ST. Self-healing and cell-free vascular grafts. Biomaterials 2025; 318:123121. [PMID: 39889339 DOI: 10.1016/j.biomaterials.2025.123121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
We developed an innovative self-healing tissue engineering vessel (SH-TEV) that heals fast after repeated needle punctures, while maintaining artery like mechanical strength and toughness even under wet conditions. The SH-TEV is designed as a bilayer tube engineered by electrospinning an autonomous self-healing polyurethane, PU-DAA, around a tube of a native biomaterial, small intestinal submucosa (SIS), that can be functionalized with biomolecules to recruit host cells and promote endothelialization. The self-healing PU-DAA was designed to incorporate multi-strength H-bonds and reversible hydrazone bonds and exhibited high strength (3.95 ± 0.16 MPa), toughness (23.01 ± 2.37 MJ/m3), and fast autonomous self-healing (86.44 ± 6.65 % after 12 h) under physiological conditions. The self-healing layer supported attachment, spreading and proliferation of fibroblasts, indicating biocompatibility. When SH-TEVs were implanted as interpositional grafts into the rat aorta for 4 weeks, they remained patent without any thrombosis (100 % animal survival and 100 % graft patency), were endothelialized and developed a smooth muscle cell containing vascular wall. In addition, they showed excellent self-healing ability following needle puncture (hemostatic time <40 s) immediately after implantation and four weeks later. Collectively, these results demonstrate the potential of SH-TEVs as vascular access conduits for hemodialysis applications.
Collapse
Affiliation(s)
- Yulun Wu
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Mohamed Alaa Mohamed
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA; Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Tai Yi
- Nationwide Children's Hospital, Columbus, OH, 43215, USA
| | - Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Clayton L Rumsey
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Martin Trebbin
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | | | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA; Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14263, USA; Center of Cell, Gene and Tissue Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA.
| |
Collapse
|
6
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025; 26:476-495. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Yang J, Sun X, Jiang H, Li J, Liang J, Lin Z. Serum-free endothelial cell culture medium for vascular smooth muscle cells sheet formation. J Biol Eng 2025; 19:51. [PMID: 40437617 PMCID: PMC12121044 DOI: 10.1186/s13036-025-00522-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 05/15/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Cell sheet technology has been identified as a promising approach for the construction of tissue-engineered vascular grafts (TEVGs). However, concerns regarding immunogenicity and ethical issues, which are raised by the use of fetal bovine serum (FBS) in traditional culture systems, limit its potential for clinical translation. Serum-free medium (SFM) has emerged as a safer and more controllable alternative, but further validation is required to determine its effectiveness and superiority in generating high-quality cell sheets. METHODS This study systematically compared cell sheets generated under SFM and 10% FBS culture conditions in terms of structure, cellular phenotype, and functional properties. The expression levels of α-SMA and SM22, markers of vascular smooth muscle cells(VSMCs), were evaluated using immunofluorescence staining, qRT-PCR, and Western blot analysis to assess cellular phenotype. Histological staining and mechanical testing were employed to compare the morphology and mechanical properties of the cell sheets, while extracellular matrix (ECM) deposition and biochemical characteristics were also analyzed. RESULTS Under SFM conditions, cells exhibited significantly higher α-SMA and SM22 expression levels (qRT-PCR showed a 1.8-fold and 2-fold increase, respectively; ****p < 0.0001) with clearer cytoskeletal arrangement. Cell sheets formed in SFM displayed comparable area(ns, p > 0.05), thickness(**p < 0.01), and mechanical properties to those cultured in 10% FBS, while ECM deposition was significantly enhanced (collagen content increased by approximately 40%, **p < 0.01). Furthermore, histological analysis revealed that cell sheets generated under SFM conditions were more compact and uniform, exhibiting superior structural organization. CONCLUSION SFM facilitates the generation of cell sheets that exhibit structural and functional properties analogous to those cultured in FBS. Additionally, SFM promotes cellular phenotype transition and ECM deposition. Consequently, SFM provides a safer, more controllable, and clinically translatable solution for cell sheet construction.
Collapse
Affiliation(s)
- Jing Yang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, 510000, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University), Guangzhou, Guangdong Province, 510000, P.R. China
| | - Xuheng Sun
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Province, 510000, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University), Guangzhou, Guangdong Province, 510000, P.R. China
- Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510000, P.R. China
| | - Hongjing Jiang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, 510000, P.R. China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University), Guangzhou, Guangdong Province, 510000, P.R. China
| | - Jiandong Li
- JIHUA Laboratory, Foshan City, Guangdong Province, 528000, P.R. China
| | - Jierong Liang
- JIHUA Laboratory, Foshan City, Guangdong Province, 528000, P.R. China
| | - Zhanyi Lin
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, 510000, P.R. China.
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University), Guangzhou, Guangdong Province, 510000, P.R. China.
| |
Collapse
|
8
|
Bai N, Li Y, Xu Q, Yang T. Revolutionizing tissue regeneration: the art of mimicking diverse collagen-based extracellular matrix hierarchies with decellularized tendon via bioskiving. Int J Biol Macromol 2025; 310:143532. [PMID: 40300687 DOI: 10.1016/j.ijbiomac.2025.143532] [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: 02/13/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Precisely simulating the collagen-based extracellular matrix (ECM) hierarchy is essential for advancing tissue regeneration. Although decellularized ECM (dECM) demonstrates considerable potential in fostering tissue regeneration by maintaining the inherent hierarchy, its application remains restricted to homologous tissues due to hierarchical variability. The current challenge involves overcoming obstacles that impede the application of dECM across diverse tissues, with particular emphasis on re-engineering one type of dECM to mimic the complex hierarchies of other tissues. The decellularized tendon, distinguished by its aligned collagen fibers, has garnered substantial interest due to its favorable structural and mechanical properties. Recently, a novel technique termed bioskiving has been developed, which uniquely disassembles macroscale decellularized tendons into microscale collagen sheets via microtome sectioning, thereby preserving micro/nano architecture of fiber alignment without chemical denaturation. These sheets can be further rotationally stacked or rolled to engineer 2D or 3D constructs replicating diverse ECM hierarchies. Bioskiving effectively dismantles the persistent hindrance that traditionally impeded the application of dECM across various tissues. This review offers an in-depth analysis of the fabrication procedure and characterization of constructs produced through bioskiving. It specifically elucidates the applications of constructs across various domains in tissue engineering, demonstrating its potential in overcoming tissue-specific constraints of dECM.
Collapse
Affiliation(s)
- Na Bai
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, School of Stomatology of Qingdao University, Qingdao 266000, Shandong, China.; Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA..
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.; Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA.
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA..
| | - Tao Yang
- Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China..
| |
Collapse
|
9
|
Choi M, Choi W, Hwang PTJ, Oh Y, Jun T, Ryu DY, Kim NK, Jang EH, Shin YR, Youn YN, Lee SH, Jung SY, Hong J. Engineered silk fibroin bio-hybrid artificial graft with releasing biological gas for enhanced circulatory stability and surgical performance. Int J Biol Macromol 2025; 309:142760. [PMID: 40185440 DOI: 10.1016/j.ijbiomac.2025.142760] [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: 01/14/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Cardiovascular disease (CVD) compromises a range of conditions affecting the heart and blood vessels, and is the leading cause of mortality globally. Vascular grafts are essential in cardiovascular surgical interventions. In clinical treatment, low mechanical durability, thrombosis and hyperplasia are primary failure modes for vascular grafts, highlighting the challenge of developing small-diameter grafts that withstand stress and integrate. A lack of suitable autologous grafts is a main cause of surgery failures. Herein, we have engineered silk fibroin (SF)-based small-diameter artificial grafts (NOeGraft) using a biologically functional polyurethane (PU) template with cost-effectiveness and high feasibility. This template facilitates the generation of biological gases via S-nitrosylation and improves mechanical properties by modulating the secondary structure of SF. Nitric oxide (NO) is one of the most essential biological gases for the cardiovascular system. NO release from NOeGraft suppresses platelet adhesion and smooth muscle cell (SMC) proliferation while scavenging reactive oxygen species (ROS) and promoting epithelial cell growth. Additionally, the suture retention strength of the NOeGraft exceeds 3.4 N. We evaluated the circulatory performance of the NOeGraft using a blood pressure-controllable system, observing no leaks or failures over 2535 min. Cost-effective NOeGraft provides biologically functional and mechanically advantageous solutions for cardiovascular surgeries.
Collapse
Affiliation(s)
- Moonhyun Choi
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Woojin Choi
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Patrick T J Hwang
- Department of Biomedical Engineering, College of Engineering, Rowan University, 201 Mullica Hill Rd., Glassboro, NJ 08028, USA
| | - Yoogyeong Oh
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Taesuk Jun
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Du Yeol Ryu
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Nam Kyun Kim
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eui Hwa Jang
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yu Rim Shin
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young-Nam Youn
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Hyun Lee
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Se Yong Jung
- Division of Pediatric Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
10
|
Imeidopf G, Khaimov D, John S, Merna N. Optimization and Standardization of Plant-Derived Vascular Scaffolds. Int J Mol Sci 2025; 26:2752. [PMID: 40141394 PMCID: PMC11942841 DOI: 10.3390/ijms26062752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Vascular graft failure rates remain unacceptably high due to thrombosis and poor integration, necessitating innovative solutions. This study optimized plant-derived extracellular matrix scaffolds as a scalable and biocompatible alternative to synthetic grafts and autologous vessels. We refined decellularization protocols to achieve >95% DNA removal while preserving mechanical properties comparable to native vessels, significantly enhancing endothelial cell seeding. Leatherleaf viburnum leaves were decellularized using sodium dodecyl sulfate-based and Trypsin/Tergitol-based treatments, achieved via clearing in bleach and Triton X-100 for 6 to 72 h. To assess the environmental influence on scaffold performance, leaves from multiple collection sites were processed using sodium dodecyl sulfate-based protocols. Scaffold performance was evaluated through tensile testing and histological analysis to assess structural integrity, while DNA quantification and endothelial cell recellularization measured biological compatibility. Sodium dodecyl sulfate-treated scaffolds with shorter clearing durations demonstrated the highest DNA removal (≥95%) while preserving mechanical properties, significantly outperforming Trypsin/Tergitol treatments. Longer clearing times reduced fiber diameter by 60%, compromising scaffold strength. Shorter clearing times preserved extracellular matrix integrity and significantly improved endothelial cell seeding efficiency. Larger leaves supported significantly higher endothelial cell densities than smaller leaves, highlighting the need for standardized material sources. Permeability tests demonstrated minimal leakage at 120 mmHg and structural stability under dynamic flow conditions, suggesting their suitability for vascular applications. These findings establish a reliable framework for optimizing plant-derived grafts, improving their reproducibility and performance for tissue engineering applications.
Collapse
Affiliation(s)
- Gianna Imeidopf
- Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY 11549, USA; (G.I.); (D.K.); (S.J.)
| | - Dara Khaimov
- Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY 11549, USA; (G.I.); (D.K.); (S.J.)
| | - Sashane John
- Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY 11549, USA; (G.I.); (D.K.); (S.J.)
- Cardiothoracic Surgery, Northwell Health, New York, NY 10022, USA
| | - Nick Merna
- Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY 11549, USA; (G.I.); (D.K.); (S.J.)
| |
Collapse
|
11
|
Zuo X, Xiao Y, Yang J, He Y, He Y, Liu K, Chen X, Guo J. Engineering collagen-based biomaterials for cardiovascular medicine. COLLAGEN AND LEATHER 2024; 6:33. [DOI: 10.1186/s42825-024-00174-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/25/2024] [Accepted: 08/21/2024] [Indexed: 01/06/2025]
Abstract
AbstractCardiovascular diseases have been the leading cause of global mortality and disability. In addition to traditional drug and surgical treatment, more and more studies investigate tissue engineering therapeutic strategies in cardiovascular medicine. Collagen interweaves in the form of trimeric chains to form the physiological network framework of the extracellular matrix of cardiac and vascular cells, possessing excellent biological properties (such as low immunogenicity and good biocompatibility) and adjustable mechanical properties, which renders it a vital tissue engineering biomaterial for the treatment of cardiovascular diseases. In recent years, promising advances have been made in the application of collagen materials in blood vessel prostheses, injectable cardiac hydrogels, cardiac patches, and hemostatic materials, although their clinical translation still faces some obstacles. Thus, we reviewed these findings and systematically summarizes the application progress as well as problems of clinical translation of collagen biomaterials in the cardiovascular field. The present review contributes to a comprehensive understanding of the application of collagen biomaterials in cardiovascular medicine.
Graphical abstract
Collapse
|
12
|
Luo X, Pang Z, Li J, Anh M, Kim BS, Gao G. Bioengineered human arterial equivalent and its applications from vascular graft to in vitro disease modeling. iScience 2024; 27:111215. [PMID: 39555400 PMCID: PMC11565542 DOI: 10.1016/j.isci.2024.111215] [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] [Indexed: 11/19/2024] Open
Abstract
Arterial disorders such as atherosclerosis, thrombosis, and aneurysm pose significant health risks, necessitating advanced interventions. Despite progress in artificial blood vessels and animal models aimed at understanding pathogenesis and developing therapies, limitations in graft functionality and species discrepancies restrict their clinical and research utility. Addressing these issues, bioengineered arterial equivalents (AEs) with enhanced vascular functions have been developed, incorporating innovative technologies that improve clinical outcomes and enhance disease progression modeling. This review offers a comprehensive overview of recent advancements in bioengineered AEs, systematically summarizing the bioengineered technologies used to construct these AEs, and discussing their implications for clinical application and pathogenesis understanding. Highlighting current breakthroughs and future perspectives, this review aims to inform and inspire ongoing research in the field, potentially transforming vascular medicine and offering new avenues for preclinical and clinical advances.
Collapse
Affiliation(s)
- Xi Luo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zherui Pang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology, Zhuhai 519088, China
| | - Minjun Anh
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
13
|
Conner AA, David D, Yim EKF. The Effects of Biomimetic Surface Topography on Vascular Cells: Implications for Vascular Conduits. Adv Healthc Mater 2024; 13:e2400335. [PMID: 38935920 DOI: 10.1002/adhm.202400335] [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: 01/28/2024] [Revised: 06/04/2024] [Indexed: 06/29/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide and represent a pressing clinical need. Vascular occlusions are the predominant cause of CVD and necessitate surgical interventions such as bypass graft surgery to replace the damaged or obstructed blood vessel with a synthetic conduit. Synthetic small-diameter vascular grafts (sSDVGs) are desired to bypass blood vessels with an inner diameter <6 mm yet have limited use due to unacceptable patency rates. The incorporation of biophysical cues such as topography onto the sSDVG biointerface can be used to mimic the cellular microenvironment and improve outcomes. In this review, the utility of surface topography in sSDVG design is discussed. First, the primary challenges that sSDVGs face and the rationale for utilizing biomimetic topography are introduced. The current literature surrounding the effects of topographical cues on vascular cell behavior in vitro is reviewed, providing insight into which features are optimal for application in sSDVGs. The results of studies that have utilized topographically-enhanced sSDVGs in vivo are evaluated. Current challenges and barriers to clinical translation are discussed. Based on the wealth of evidence detailed here, substrate topography offers enormous potential to improve the outcome of sSDVGs and provide therapeutic solutions for CVDs.
Collapse
Affiliation(s)
- Abigail A Conner
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Dency David
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - 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
|
14
|
Chernonosova VS, Osipova OS, Nuankai Z, Shundrina IK, Murashov IS, Larichev YV, Karpenko AA, Laktionov PP. Evaluation of properties for Carbothane™ 3575A-based electrospun vascular grafts in vitroand in vivo. Biomed Mater 2024; 19:065012. [PMID: 39255825 DOI: 10.1088/1748-605x/ad792d] [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: 04/13/2024] [Accepted: 09/10/2024] [Indexed: 09/12/2024]
Abstract
Bioengineered vascular grafts (VGs) have emerged as a promising alternative to the treatment of damaged or occlusive vessels. It is thought that polyurethane (PU)-based scaffolds possess suitable hemocompatibility and biomechanics comparable to those of normal blood vessels. In this study, we investigated the properties of electrospun scaffolds comprising various blends of biostable polycarbonate-based PU (Carbothane™ 3575A) and gelatin. Scaffolds were characterized by scanning electron microscopy, infra-red spectroscopy, small-angle x-ray scattering, stress-loading tests, and interactions with primary human cells and blood. Data fromin vitroexperiments demonstrated that a scaffold produced from a blend of 5% Carbothane™ 3575A and 10% gelatin has proven to be a suitable material for fabricating a small-diameter VG. A comparativein vivostudy of such VGs and expanded polytetrafluoroethylene (ePTFE) grafts implanted in the abdominal aorta of Wistar rats was performed. The data of intravital study and histological examination indicated that Carbothane-based electrospun grafts outclass ePTFE grafts and represent a promising device for preclinical studies to satisfy vascular surgery needs.
Collapse
Affiliation(s)
- Vera S Chernonosova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Olesia S Osipova
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Zhou Nuankai
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Inna K Shundrina
- Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Ivan S Murashov
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Yurii V Larichev
- Boreskov Institute of Catalysis, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Andrey A Karpenko
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Pavel P Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
15
|
Franko R, de Almeida Monteiro Melo Ferraz M. Exploring the potential of in vitro extracellular vesicle generation in reproductive biology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70007. [PMID: 39238549 PMCID: PMC11375532 DOI: 10.1002/jex2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
The interest in the growing field of extracellular vesicle (EV) research highlights their significance in intercellular signalling and the selective transfer of biological information between donor and recipient cells. EV studies have provided valuable insights into intercellular communication mechanisms, signal identification and their involvement in disease states, offering potential avenues for manipulating pathological conditions, detecting biomarkers and developing drug-delivery systems. While our understanding of EV functions in reproductive tissues has significantly progressed, exploring their potential as biomarkers for infertility, therapeutic interventions and enhancements in assisted reproductive technologies remains to be investigated. This knowledge gap stems partly from the difficulties associated with large-scale EV production relevant to clinical applications. Most existing studies on EV production rely on conventional 2D cell culture systems, characterized by suboptimal EV yields and a failure to replicate in vivo conditions. This results in the generation of EVs that differ from their in vivo counterparts. Hence, this review firstly delves into the importance of EVs in reproduction to then expand on current techniques for in vitro EV production, specifically examining diverse methods of culture and the potential of bioengineering technologies to establish innovative systems for enhanced EV production.
Collapse
Affiliation(s)
- Roksan Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| | - Marcia de Almeida Monteiro Melo Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| |
Collapse
|
16
|
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
|
17
|
Snyder Y, Jana S. Influence of Substrate Structure and Associated Properties on Endothelial Cell Behavior in the Context of Behaviors Associated with Laminar Flow Conditions. ACS APPLIED BIO MATERIALS 2024; 7:4664-4678. [PMID: 38939951 DOI: 10.1021/acsabm.4c00504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
In order to treat most vascular diseases, arterial grafts are commonly employed for replacing small-diameter vessels, yet they often cause thrombosis. The growth of endothelial cells along the interior surfaces of these grafts (substrates) is critical to mitigate thrombosis. Typically, endothelial cells are cultured inside these grafts under laminar flow conditions to emulate the native environment of blood vessels and produce an endothelium. Alternatively, the substrate structure could have a similar influence on endothelial cell behavior as laminar flow conditions. In this study, we investigated whether substrates with aligned fiber structures could induce responses in human umbilical vein endothelial cells (HUVECs) akin to those elicited by laminar flow. Our observations revealed that HUVECs on aligned substrates displayed significant morphological changes, aligning parallel to the fibers, similar to effects reported under laminar flow conditions. Conversely, HUVECs on random substrates maintained their characteristic cobblestone appearance. Notably, cell migration was more significant on aligned substrates. Also, we observed that while vWF expression was similar between both substrates, the HUVECs on aligned substrates showed more expression of platelet/endothelial cell adhesion molecule-1 (PECAM-1/CD31), laminin, and collagen IV. Additionally, these cells exhibited increased gene expression related to critical functions such as proliferation, extracellular matrix production, cytoskeletal reorganization, autophagy, and antithrombotic activity. These findings indicated that aligned substrates enhanced endothelial growth and behavior compared to random substrates. These improvements are similar to the beneficial effects of laminar flow on endothelial cells, which are well-documented compared to static or turbulent flow conditions.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65211, United States
| | - Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
18
|
Li J, Lan T, Guo Q, Zhang C, Lu X, Hu X, Shen X, Zhang Y. Mitochondria-Targeted Natural Antioxidant Nanosystem for Diabetic Vascular Calcification Therapy. Biomacromolecules 2024; 25:4329-4343. [PMID: 38833553 DOI: 10.1021/acs.biomac.4c00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The development of nanotherapy targeting mitochondria to alleviate oxidative stress is a critical therapeutic strategy for vascular calcification (VC) in diabetes. In this study, we engineered mitochondria-targeted nanodrugs (T4O@TPP/PEG-PLGA) utilizing terpinen-4-ol (T4O) as a natural antioxidant and mitochondrial protector, PEG-PLGA as the nanocarrier, and triphenylphosphine (TPP) as the mitochondrial targeting ligand. In vitro assessments demonstrated enhanced cellular uptake of T4O@TPP/PEG-PLGA, with effective mitochondrial targeting. This nanodrug successfully reduced oxidative stress induced by high glucose levels in vascular smooth muscle cells. In vivo studies showed prolonged retention of the nanomaterials in the thoracic aorta for up to 24 h. Importantly, experiments in diabetic VC models underscored the potent antioxidant properties of T4O@TPP/PEG-PLGA, as evidenced by its ability to mitigate VC and restore mitochondrial morphology. These results suggest that these nanodrugs could be a promising strategy for managing diabetic VC.
Collapse
Affiliation(s)
- Jinjin Li
- The Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Tianyu Lan
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- College of Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, Guizhou, China
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Chuang Zhang
- The Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Xin Lu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Xiaoxia Hu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Yanyan Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| |
Collapse
|
19
|
Vuong TNAM, Bartolf‐Kopp M, Andelovic K, Jungst T, Farbehi N, Wise SG, Hayward C, Stevens MC, Rnjak‐Kovacina J. Integrating Computational and Biological Hemodynamic Approaches to Improve Modeling of Atherosclerotic Arteries. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307627. [PMID: 38704690 PMCID: PMC11234431 DOI: 10.1002/advs.202307627] [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: 10/12/2023] [Revised: 03/12/2024] [Indexed: 05/07/2024]
Abstract
Atherosclerosis is the primary cause of cardiovascular disease, resulting in mortality, elevated healthcare costs, diminished productivity, and reduced quality of life for individuals and their communities. This is exacerbated by the limited understanding of its underlying causes and limitations in current therapeutic interventions, highlighting the need for sophisticated models of atherosclerosis. This review critically evaluates the computational and biological models of atherosclerosis, focusing on the study of hemodynamics in atherosclerotic coronary arteries. Computational models account for the geometrical complexities and hemodynamics of the blood vessels and stenoses, but they fail to capture the complex biological processes involved in atherosclerosis. Different in vitro and in vivo biological models can capture aspects of the biological complexity of healthy and stenosed vessels, but rarely mimic the human anatomy and physiological hemodynamics, and require significantly more time, cost, and resources. Therefore, emerging strategies are examined that integrate computational and biological models, and the potential of advances in imaging, biofabrication, and machine learning is explored in developing more effective models of atherosclerosis.
Collapse
Affiliation(s)
| | - Michael Bartolf‐Kopp
- Department of Functional Materials in Medicine and DentistryInstitute of Functional Materials and Biofabrication (IFB)KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)University of WürzburgPleicherwall 297070WürzburgGermany
| | - Kristina Andelovic
- Department of Functional Materials in Medicine and DentistryInstitute of Functional Materials and Biofabrication (IFB)KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)University of WürzburgPleicherwall 297070WürzburgGermany
| | - Tomasz Jungst
- Department of Functional Materials in Medicine and DentistryInstitute of Functional Materials and Biofabrication (IFB)KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)University of WürzburgPleicherwall 297070WürzburgGermany
- Department of Orthopedics, Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht3584Netherlands
| | - Nona Farbehi
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydney2052Australia
- Tyree Institute of Health EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Garvan Weizmann Center for Cellular GenomicsGarvan Institute of Medical ResearchSydneyNSW2010Australia
| | - Steven G. Wise
- School of Medical SciencesUniversity of SydneySydneyNSW2006Australia
| | - Christopher Hayward
- St Vincent's HospitalSydneyVictor Chang Cardiac Research InstituteSydney2010Australia
| | | | - Jelena Rnjak‐Kovacina
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydney2052Australia
- Tyree Institute of Health EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicine (ACN)University of New South WalesSydneyNSW2052Australia
| |
Collapse
|
20
|
Wang X, Fu H, Wu H, Peng X, Peng X, Yu X, Liu H, Wu J, Luo L, Yan S, Cheng X, Zhou X, Yuan X. Corilagin functionalized decellularized extracellular matrix as artificial blood vessels with improved endothelialization and anti-inflammation by reactive oxygen species scavenging. Regen Biomater 2024; 11:rbae074. [PMID: 39664941 PMCID: PMC11631697 DOI: 10.1093/rb/rbae074] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 12/13/2024] Open
Abstract
The performance of biological-originated blood vessels in clinical remains disappointing due to fast occlusion caused by acute thrombosis or long-standing inflammation. How to prevent rapid degradation and inhibit acute inflammation but maintain their high bioactivity is still a significant challenge. As a bioactive polyphenol in various traditional Chinese medicine, Corilagin (Cor) exhibits excellent anticoagulant, anti-inflammatory and rapid ROS consumption properties. Inspired by abundant supramolecular interactions in organisms, we selected it to crosslink tissues via purely H-bonds to simulate these natural interactions without introducing potential toxic aldehyde or carboxyl groups. Results show that 2 mg/ml was selected as the optimal Cor concentration to form a stable crosslinking network (FI > 95%) and effectively delay their degradation. Cor modification not only enhances ECs adhesion and monolayer function via accelerating VEGF and TGF-β secretion but also promotes macrophage transformation from pro-inflammatory M1 phenotype to anti-inflammatory M2 ones. In vitro and ex-vivo studies implied that Cor-crosslinked samples exhibited low platelet accumulation and decreased thrombin generation. In vivo evaluation further confirmed that Cor-introducing could effectively consume ROS, thus exhibiting rapid endothelialization, suppressed inflammation and reduced mineral deposition. Overall, Cor crosslinking provided a bright future for blood vessels' long-term patency and adapted to various blood-contacting surfaces.
Collapse
Affiliation(s)
- Xu Wang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Hanmei Fu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Huibin Wu
- College of Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Xiaohua Peng
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xu Peng
- College of Polymer Science and Engineering & Laboratory Animal Center, Sichuan University, Chengdu 610065, China
| | - Xixun Yu
- College of Polymer Science and Engineering & Laboratory Animal Center, Sichuan University, Chengdu 610065, China
| | - Hui Liu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Junmei Wu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- College of Traditional Chinese Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ling Luo
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shan Yan
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xinglin Cheng
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiong Zhou
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Xiangyang Yuan
- College of Science, Sichuan Agricultural University, Ya’an 625014, China
| |
Collapse
|
21
|
Hernandez-Sanchez D, Comtois-Bona M, Muñoz M, Ruel M, Suuronen EJ, Alarcon EI. Manufacturing and validation of small-diameter vascular grafts: A mini review. iScience 2024; 27:109845. [PMID: 38799581 PMCID: PMC11126982 DOI: 10.1016/j.isci.2024.109845] [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] [Indexed: 05/29/2024] Open
Abstract
The field of small-diameter vascular grafts remains a challenge for biomaterials scientists. While decades of research have brought us much closer to developing biomimetic materials for regenerating tissues and organs, the physiological challenges involved in manufacturing small conduits that can transport blood while not inducing an immune response or promoting blood clots continue to limit progress in this area. In this short review, we present some of the most recent methods and advancements made by researchers working in the field of small-diameter vascular grafts. We also discuss some of the most critical aspects biomaterials scientists should consider when developing lab-made small-diameter vascular grafts.
Collapse
Affiliation(s)
- Deyanira Hernandez-Sanchez
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Maxime Comtois-Bona
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Marcelo Muñoz
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Marc Ruel
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, 451 Smyth Road, Ottawa ON K1H8M5, Canada
| | - Erik J. Suuronen
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, 451 Smyth Road, Ottawa ON K1H8M5, Canada
| | - Emilio I. Alarcon
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H8M5, Canada
| |
Collapse
|
22
|
Zhao Y, Peng H, Sun L, Tong J, Cui C, Bai Z, Yan J, Qin D, Liu Y, Wang J, Wu X, Li B. The application of small intestinal submucosa in tissue regeneration. Mater Today Bio 2024; 26:101032. [PMID: 38533376 PMCID: PMC10963656 DOI: 10.1016/j.mtbio.2024.101032] [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: 12/13/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
The distinctive three-dimensional architecture, biological functionality, minimal immunogenicity, and inherent biodegradability of small intestinal submucosa extracellular matrix materials have attracted considerable interest and found wide-ranging applications in the domain of tissue regeneration engineering. This article presents a comprehensive examination of the structure and role of small intestinal submucosa, delving into diverse preparation techniques and classifications. Additionally, it proposes approaches for evaluating and modifying SIS scaffolds. Moreover, the advancements of SIS in the regeneration of skin, bone, heart valves, blood vessels, bladder, uterus, and urethra are thoroughly explored, accompanied by their respective future prospects. Consequently, this review enhances our understanding of the applications of SIS in tissue and organ repair and keeps researchers up-to-date with the latest research advancements in this area.
Collapse
Affiliation(s)
- Yifan Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Hongyi Peng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Lingxiang Sun
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Jiahui Tong
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Chenying Cui
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Ziyang Bai
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Jingyu Yan
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Danlei Qin
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Yingyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Jue Wang
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| |
Collapse
|
23
|
Li Z, Giarto J, Zhang J, Gim J, Chen E, Enriquez E, Jafuta L, Mahalingam E, Turng LS. Design and Synthesis of P(AAm-co-NaAMPS)-Alginate-Xanthan Hydrogels and the Study of Their Mechanical and Rheological Properties in Artificial Vascular Graft Applications. Gels 2024; 10:319. [PMID: 38786235 PMCID: PMC11121731 DOI: 10.3390/gels10050319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the number one cause of mortality among non-communicable diseases worldwide. Expanded polytetrafluoroethylene (ePTFE) is a widely used material for making artificial vascular grafts to treat CVDs; however, its application in small-diameter vascular grafts is limited by the issues of thrombosis formation and intimal hyperplasia. This paper presents a novel approach that integrates a hydrogel layer on the lumen of ePTFE vascular grafts through mechanical interlocking to efficiently facilitate endothelialization and alleviate thrombosis and restenosis problems. This study investigated how various gel synthesis variables, including N,N'-Methylenebisacrylamide (MBAA), sodium alginate, and calcium sulfate (CaSO4), influence the mechanical and rheological properties of P(AAm-co-NaAMPS)-alginate-xanthan hydrogels intended for vascular graft applications. The findings obtained can provide valuable guidance for crafting hydrogels suitable for artificial vascular graft fabrication. The increased sodium alginate content leads to increased equilibrium swelling ratios, greater viscosity in hydrogel precursor solutions, and reduced transparency. Adding more CaSO4 decreases the swelling ratio of a hydrogel system, which offsets the increased swelling ratio caused by alginate. Increased MBAA in the hydrogel system enhances both the shear modulus and Young's modulus while reducing the transparency of the hydrogel system and the pore size of freeze-dried samples. Overall, Hydrogel (6A12M) with 2.58 mg/mL CaSO4 was the optimal candidate for ePTFE-hydrogel vascular graft applications due to its smallest pore size, highest shear storage modulus and Young's modulus, smallest swelling ratio, and a desirable precursor solution viscosity that facilitates fabrication.
Collapse
Affiliation(s)
- Zhutong Li
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (Z.L.); (E.C.); (E.E.); (L.J.)
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; (J.G.); (E.M.)
| | - Joshua Giarto
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; (J.G.); (E.M.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI 53706, USA
| | - Jue Zhang
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53715, USA;
| | - Jinsu Gim
- Dongnam Division, Korea Institute of Industrial Technology (KITECH), Jinju 52845, Republic of Korea;
| | - Edward Chen
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (Z.L.); (E.C.); (E.E.); (L.J.)
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; (J.G.); (E.M.)
| | - Eduardo Enriquez
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (Z.L.); (E.C.); (E.E.); (L.J.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI 53706, USA
| | - Lauren Jafuta
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (Z.L.); (E.C.); (E.E.); (L.J.)
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; (J.G.); (E.M.)
| | - Esha Mahalingam
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; (J.G.); (E.M.)
- College of Letters and Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lih-Sheng Turng
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; (Z.L.); (E.C.); (E.E.); (L.J.)
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; (J.G.); (E.M.)
| |
Collapse
|
24
|
Kawcher Alam M, Sahadat Hossain M, Anisur Rahman Dayan M, Bahadur NM, Shaikh MAA, Ahmed S. Fabrication and Characterization of a Bioscaffold Using Hydroxyapatite and Unsaturated Polyester Resin. ACS OMEGA 2024; 9:15210-15221. [PMID: 38585056 PMCID: PMC10993257 DOI: 10.1021/acsomega.3c09599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 04/09/2024]
Abstract
Outstanding biodegradability and biocompatibility are attributes associated with particular polyester substances that make this group useful in specific biomedical fields. To assess the potential as a biomaterial, a novel composite consisting of hydroxyapatite (HAp) and unsaturated polyester resin (UPR) was developed in this work. Using a hand-lay-up technique, various percentages (50, 40, 30, 20, and 10%) of HAp were reinforced into the UPR matrix to fabricate composite materials out of glass sheets. Prior to processing of the composite samples, hydroxyapatite was chemically synthesized in a wet chemical manner. Using a universal testing machine (UTM), Fourier transform infrared (FTIR) spectroscopy, scanning electron microscopy (SEM), and thermo-gravimetric analysis (TGA), the fabricated samples were characterized. The crystallographic parameters of synthesized hydroxyapatite (HAp) were also estimated through a range of formulas. The optimal amount for hydroxyapatite was 40% according to the findings of the tensile strength (TS), tensile modulus (TM), percentage of elongation at break (EB), bending strength (BS), and bending modulus (BM). Improvements in TS, TM, BS, and BM for the ideal combination were 39.39, 9.21, 912.05, and 259.96%, in each case, over the controlled one. Thermogravimetric analysis (TGA) has been implemented to determine the degradation temperature of the fabricated composites up to 600 °C.
Collapse
Affiliation(s)
- Md. Kawcher Alam
- Glass
Research Division, Institute of Glass & Ceramic Research and Testing, Bangladesh Council of Scientific and Industrial Research
(BCSIR), Dhaka 1205, Bangladesh
- Department
of Applied Chemistry and Chemical Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md. Sahadat Hossain
- Glass
Research Division, Institute of Glass & Ceramic Research and Testing, Bangladesh Council of Scientific and Industrial Research
(BCSIR), Dhaka 1205, Bangladesh
| | - Md. Anisur Rahman Dayan
- Textile
Physics Division, Bangladesh Jute Research
Institute, Manik Mia
Avenue, Dhaka 1207, Bangladesh
| | - Newaz Mohammed Bahadur
- Department
of Applied Chemistry and Chemical Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Md. Aftab Ali Shaikh
- Glass
Research Division, Institute of Glass & Ceramic Research and Testing, Bangladesh Council of Scientific and Industrial Research
(BCSIR), Dhaka 1205, Bangladesh
- Department
of Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Samina Ahmed
- Glass
Research Division, Institute of Glass & Ceramic Research and Testing, Bangladesh Council of Scientific and Industrial Research
(BCSIR), Dhaka 1205, Bangladesh
- BCSIR
Dhaka Laboratories, Bangladesh Council of
Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| |
Collapse
|
25
|
Lee DU, Kayumov M, Park J, Park SK, Kang Y, Ahn Y, Kim W, Yoo SH, Park JK, Kim BG, Oh YS, Jeong IS, Choi DY. Antibiofilm and antithrombotic hydrogel coating based on superhydrophilic zwitterionic carboxymethyl chitosan for blood-contacting devices. Bioact Mater 2024; 34:112-124. [PMID: 38204564 PMCID: PMC10777421 DOI: 10.1016/j.bioactmat.2023.12.009] [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: 07/30/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Blood-contacting devices must be designed to minimize the risk of bloodstream-associated infections, thrombosis, and intimal lesions caused by surface friction. However, achieving effective prevention of both bloodstream-associated infections and thrombosis poses a challenge due to the conflicting nature of antibacterial and antithrombotic activities, specifically regarding electrostatic interactions. This study introduced a novel biocompatible hydrogel of sodium alginate and zwitterionic carboxymethyl chitosan (ZW@CMC) with antibacterial and antithrombotic activities for use in catheters. The ZW@CMC hydrogel demonstrates a superhydrophilic surface and good hygroscopic properties, which facilitate the formation of a stable hydration layer with low friction. The zwitterionic-functionalized CMC incorporates an additional negative sulfone group and increased negative charge density in the carboxyl group. This augmentation enhances electrostatic repulsion and facilitates the formation of hydration layer. This leads to exceptional prevention of blood clotting factor adhesion and inhibition of biofilm formation. Subsequently, the ZW@CMC hydrogel exhibited biocompatibility with tests of in vitro cytotoxicity, hemolysis, and catheter friction. Furthermore, in vivo tests of antithrombotic and systemic inflammation models with catheterization indicated that ZW@CMC has significant advantages for practical applications in cardiovascular-related and sepsis treatment. This study opens a new avenue for the development of chitosan-based multifunctional hydrogel for applications in blood-contacting devices.
Collapse
Affiliation(s)
- Dong Uk Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon, 38822, Republic of Korea
| | - Mukhammad Kayumov
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Junghun Park
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon, 38822, Republic of Korea
| | - Se Kye Park
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon, 38822, Republic of Korea
| | - Yeongkwon Kang
- Research Institute of Advanced Materials, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yejin Ahn
- Department of Organic and Nano System Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Woojin Kim
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon, 38822, Republic of Korea
| | - Seung Hwa Yoo
- Department of Quantum System Engineering, Jeonbuk National University, Jeonju-si, 54896, Republic of Korea
| | | | - Bong-Gi Kim
- Department of Organic and Nano System Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Yong Suk Oh
- Department of Mechanical Engineering, Changwon National University, Changwon, 51140, Republic of Korea
| | - In-Seok Jeong
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Dong Yun Choi
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon, 38822, Republic of Korea
| |
Collapse
|
26
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
27
|
Kaneko J, Hayashi Y, Kazami Y, Nishioka Y, Miyata A, Ichida A, Kawaguchi Y, Akamatsu N, Hasegawa K. Resection and reconstruction of the largest abdominal vein system (the inferior vena cava, hepatic, and portal vein): a narrative review. Transl Gastroenterol Hepatol 2024; 9:23. [PMID: 38716218 PMCID: PMC11074493 DOI: 10.21037/tgh-23-90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/10/2024] [Indexed: 02/12/2025] Open
Abstract
BACKGROUND AND OBJECTIVE As tumors invade major abdominal veins, surgical procedures are transformed from simple and basic to complicated and challenging. In this narrative review, we focus on what is currently known and not known regarding the technical aspects of major abdominal venous resection and its reconstruction, patency, and oncologic benefit in a cross-cutting perspective. METHODS A systematic literature search was performed in PubMed and Semantic Scholar from inception up to October 18, 2023. We reviewed 106 papers by title, abstract, and full text regarding resection or reconstruction of the inferior vena cava, hepatic vein confluence, portal vein (PV), and middle hepatic vein (MHV) tributaries in living donor liver transplantation (LDLT) in a cross-cutting perspective. KEY CONTENT AND FINDINGS The oncologic benefit of aggressive hepatic vein resection with suitable reconstruction against adenocarcinoma remains unclear, and further studies are required to clarify this point. A superior mesenteric/PV resection is now a universal, indispensable, and effective procedure for pancreatic ductal adenocarcinoma. Although many case series using tailor-made autologous venous grafts have been reported, not only size mismatch but also additional surgical incisions and a longer operation time remain obstacles for venous reconstruction. The use of autologous alternative tissue remains only an alternative procedure because the patency rate of customized tubular conduit type to interpose or replace the resected vein is not known. Unlike arterial replacement, venous replacement using synthetic vascular grafts is still rarely reported and there are several inherent limitations except for reconstruction of tributaries of MHV in LDLT. CONCLUSIONS Various approaches to abdominal vein resection and replacement or reconstruction are technically feasible with satisfactory results. Synthetic vascular grafts may be appropriate but have a certain rate of complications.
Collapse
Affiliation(s)
- Junichi Kaneko
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Hayashi
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Kazami
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yujiro Nishioka
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akinori Miyata
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiko Ichida
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshikuni Kawaguchi
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhisa Akamatsu
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery, Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Zhang W, Fukazawa K, Mahara A, Jiang H, Yamaoka T. Photo-induced universal modification of small-diameter decellularized blood vessels with a hemocompatible peptide improves in vivo patency. Acta Biomater 2024; 176:116-127. [PMID: 38232911 DOI: 10.1016/j.actbio.2024.01.012] [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: 07/14/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
Decellularized vessels (DVs) have the potential to serve as available grafts for small-diameter vascular (<6 mm) reconstruction. However, the absence of functional endothelia makes them likely to trigger platelet aggregation and thrombosis. Luminal surface modification is an efficient approach to prevent thrombosis and promote endothelialization. Previously, we identified a hemocompatible peptide, HGGVRLY, that showed endothelial affinity and antiplatelet ability. By conjugating HGGVRLY with a phenylazide group, we generated a photoreactive peptide that can be modified onto multiple materials, including non-denatured extracellular matrices. To preserve the natural collagen of DVs as much as possible, we used a lower ultrahydrostatic pressure than that previously reported to prepare decellularized grafts. The photoreactive HGGVRLY peptide could be modified onto DV grafts via UV exposure for only 2 min. Modified DVs showed improved endothelial affinity and antiplatelet ability in vitro. When rat abdominal aortas were replaced with DVs, modified DVs with more natural collagen demonstrated the highest patent rate after 10 weeks. Moreover, the photoreactive peptide remained on the lumen surface of DVs over two months after implantation. Therefore, the photoreactive peptide could be efficiently and sustainably modified onto DVs with more natural collagens, resulting in improved hemocompatibility. STATEMENT OF SIGNIFICANCE: We employed a relatively lower ultrahydrostatic pressure to prepare decellularized vessels (DVs) with less denatured collagens to provide a more favorable environment for cell migration and proliferation. The hemocompatibility of DV luminal surface can be enhanced by peptide modification, but undenatured collagens are difficult to modify. We innovatively introduce a phenylazide group into the hemocompatible peptide HGGVRLY, which we previously identified to possess endothelial affinity and antiplatelet ability, to generate a photoreactive peptide. The photoreactive peptide can be efficiently and stably modified onto DVs with more natural collagens. DV grafts modified with photoreactive peptide exhibit enhanced in vivo patency. Furthermore, the sustainability of photoreactive peptide modification on DV grafts within bloodstream is evident after two months of transplantation.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan; Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing China
| | - Kyoko Fukazawa
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Haiyue Jiang
- Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing China
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan.
| |
Collapse
|
29
|
Sun D, Katare R, Sethu P, Cheng P, Fan Y. Editorial: Constructing the vascular or cardiac tissue and organoids: the combination of biomedicine and engineering. Front Cardiovasc Med 2024; 11:1371074. [PMID: 38433752 PMCID: PMC10904631 DOI: 10.3389/fcvm.2024.1371074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Affiliation(s)
- Dayu Sun
- Department of Anatomy, Third Military Medical University, Chongqing, China
| | - Rajesh Katare
- Department of Physiology, HeartOtago, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Palaniappan Sethu
- Division of Cardiovascular Disease, Department of Medicine and Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yonghong Fan
- Laboratory of Basic Medicine, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
30
|
Liu W, Wang X. Recent Advances of Nanogenerator Technology for Cardiovascular Sensing and Monitoring. NANO ENERGY 2023; 117:108910. [PMID: 39183759 PMCID: PMC11343574 DOI: 10.1016/j.nanoen.2023.108910] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Cardiovascular sensing and monitoring is a widely used function in cardiovascular devices. Nowadays, achieving desired flexibility, wearability and implantability becomes a major design goal for the advancement of this family of devices. As an emerging technology, nanogenerator (NG) offers an intriguing promise for replacing the battery, an essential obstacle toward tissue-like soft electronics. This article reviews most recent advancements in NG technology for advanced cardiovascular sensing and monitoring. Based on the application targets, the discuss covers implantable NGs on hearts, implantable NGs for blood vessel grafts and patches, and wearable NGs with various sensing functions. The applications of NGs as a power source and as an electromechanical sensing element are both discussed. At the end, current challenges in this direction and future research perspectives are elaborated. This emerging and impactful application direction reviewed in this article is expected to inspire many new research and commercialization opportunities in the field of NG technology.
Collapse
Affiliation(s)
- Wenjian Liu
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xudong Wang
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
31
|
Li Z, Giarto J, Zhang J, Kulkarni N, Mahalingam E, Klipstine W, Turng LS. Anti-thrombotic poly(AAm-co-NaAMPS)-xanthan hydrogel-expanded polytetrafluoroethylene (ePTFE) vascular grafts with enhanced endothelialization and hemocompatibility properties. BIOMATERIALS ADVANCES 2023; 154:213625. [PMID: 37722163 PMCID: PMC10841274 DOI: 10.1016/j.bioadv.2023.213625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death among all non-communicable diseases globally. Although expanded polytetrafluoroethylene (ePTFE) has been widely used for larger-diameter vascular graft transplantation, the persistent thrombus formation and intimal hyperplasia of small-diameter vascular grafts (SDVGs) made of ePTFE to treat severe CVDs remain the biggest challenges due to lack of biocompatibility and endothelium. In this study, bi-layered poly(acrylamide-co-2-Acrylamido-2-methyl-1-propanesulfonic acid sodium)-xanthan hydrogel-ePTFE (poly(AAm-co-NaAMPS)-xanthan hydrogel-ePTFE) vascular grafts capable of promoting endothelialization and prohibiting thrombosis were synthesized and fabricated. While the external ePTFE layer of the vascular grafts provided the mechanical stability, the inner hydrogel layer offered much-needed cytocompatibility, hemocompatibility, and endothelialization functions. The interface morphology between the inner hydrogel layer and the outer ePTFE layer was observed by scanning electron microscope (SEM), which revealed that the hydrogel was well attached to the porous ePTFE through mechanical interlocking. Among all the hydrogel compositions tested with cell culture using human umbilical vein endothelial cells (HUVECs), the hydrogel with the molar ratio of 40:60 (NaAMPS/AAm) composition (i.e., Hydrogel 40:60) exhibited the best endothelialization function, as it produced the largest endothelialization area that was three times more than of that of plain ePTFE on day 14, maintained the highest average cell viability, and had the best cell morphology. Hydrogel 40:60 also showed excellent hemocompatibility, prolonged activated partial thromboplastin time (aPTT), and good mechanical properties. Overall, bi-layered poly(AAm-co-NaAMPS)-xanthan hydrogel-ePTFE vascular grafts with the Hydrogel 40:60 composition could potentially solve the critical challenge of thrombus formation in vascular graft transplantation applications.
Collapse
Affiliation(s)
- Zhutong Li
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Joshua Giarto
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jue Zhang
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Neha Kulkarni
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Esha Mahalingam
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; College of Letters and Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Will Klipstine
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Lih-Sheng Turng
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Mechanical Engineering, Chang Gung University, Tao-Yuan 33302, Taiwan.
| |
Collapse
|
32
|
Yachmaneni A, Jajoo S, Mahakalkar C, Kshirsagar S, Dhole S. A Comprehensive Review of the Vascular Consequences of Diabetes in the Lower Extremities: Current Approaches to Management and Evaluation of Clinical Outcomes. Cureus 2023; 15:e47525. [PMID: 38022307 PMCID: PMC10664734 DOI: 10.7759/cureus.47525] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Diabetes mellitus is a global health concern characterized by chronic hyperglycemia, and its vascular consequences in the lower extremities pose significant challenges for individuals living with the condition. This comprehensive review delves into the multifaceted landscape of diabetes-related vascular complications in the lower limbs, with a primary focus on current strategies for management and the evaluation of clinical outcomes. This review achieves several critical objectives by synthesizing existing knowledge and research findings. It elucidates the intricate pathophysiological mechanisms underpinning these complications, shedding light on the cellular and molecular processes involved. Additionally, it outlines clinical assessment and diagnostic strategies used to identify and stratify risk, ranging from cutting-edge imaging techniques to clinical examinations. The review comprehensively examines current management strategies, encompassing lifestyle modifications, pharmacological interventions, surgical procedures, and wound care practices. Moreover, it assesses and analyzes clinical outcomes, including limb salvage rates, amputation rates, and overall quality of life for individuals undergoing treatment. In addressing the challenges faced in managing these complications, this review aims to contribute to improved patient care. It proposes future research directions to enhance the management and outcomes of diabetes-related vascular consequences in the lower extremities.
Collapse
Affiliation(s)
- Akanksha Yachmaneni
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Suhas Jajoo
- Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Chandrashekhar Mahakalkar
- Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Shivani Kshirsagar
- Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Simran Dhole
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
33
|
Li S, Zhao F, Tang Y, Zhang Y, Rong H, Liu L, Gao R, Liu X, Huangfu Y, Bai Y, Feng Z, Guo Z, Dong A, Wang W, Kong D, Huang P. Bioinspired, Anticoagulative, 19 F MRI-Visualizable Bilayer Hydrogel Tubes as High Patency Small-Diameter Vascular Grafts. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302621. [PMID: 37340585 DOI: 10.1002/smll.202302621] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/29/2023] [Indexed: 06/22/2023]
Abstract
The clinical patency of small-diameter vascular grafts (SDVGs) (ID < 6 mm) is limited, with the formation of mural thrombi being a major threat of this limitation. Herein, a bilayered hydrogel tube based on the essential structure of native blood vessels is developed by optimizing the relation between vascular functions and the molecular structure of hydrogels. The inner layer of the SDVGs comprises a zwitterionic fluorinated hydrogel, avoiding the formation of thromboinflammation-induced mural thrombi. Furthermore, the position and morphology of the SDVGs can be visualized via 19 F/1 H magnetic resonance imaging. The outer poly(N-acryloyl glycinamide) hydrogel layer of SDVGs provides matched mechanical properties with native blood vessels through the multiple and controllable intermolecular hydrogen-bond interactions, which can withstand the accelerated fatigue test under pulsatile radial pressure for 380 million cycles (equal to a service life of 10 years in vivo). Consequently, the SDVGs exhibit higher patency (100%) and more stable morphology following porcine carotid artery transplantation for 9 months and rabbit carotid artery transplantation for 3 months. Therefore, such a bioinspired, antithrombotic, and visualizable SDVG presents a promising design approach for long-term patency products and great potential of helping patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Shuangyang Li
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Feng Zhao
- Chest hospital, Tianjin University, Tianjin, 300222, China
| | - Yipeng Tang
- Chest hospital, Tianjin University, Tianjin, 300222, China
| | - Yiqun Zhang
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Hui Rong
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Lingyuan Liu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Rui Gao
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Xiang Liu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Yini Huangfu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Yunpeng Bai
- Chest hospital, Tianjin University, Tianjin, 300222, China
| | - Zujian Feng
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Zhigang Guo
- Chest hospital, Tianjin University, Tianjin, 300222, China
| | - Anjie Dong
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering(MOE), Tianjin University, Tianjin, 300072, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
34
|
Kirkton RD, Watson JDB, Houston R, Prichard HL, Niklason LE, Rasmussen TE. Evaluation of vascular repair by tissue-engineered human acellular vessels or expanded polytetrafluoroethylene grafts in a porcine model of limb ischemia and reperfusion. J Trauma Acute Care Surg 2023; 95:234-241. [PMID: 36943014 PMCID: PMC10389495 DOI: 10.1097/ta.0000000000003974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/24/2023] [Accepted: 03/12/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND This study evaluated performance of a tissue-engineered human acellular vessel (HAV) in a porcine model of acute vascular injury and ischemia. The HAV is an engineered blood vessel consisted of human vascular extracellular matrix proteins. Limb reperfusion and vascular outcomes of the HAV were compared with those from synthetic expanded polytetrafluoroethylene (ePTFE) grafts. METHODS Thirty-six pigs were randomly assigned to four treatment groups, receiving either the HAV or a PTFE graft following a hind limb ischemia period of either 0 or 6 hours. All grafts were 3-cm-long interposition 6-mm diameter grafts implanted within the right iliac artery. Animals were not immunosuppressed and followed for up to 28 days after surgery. Assessments performed preoperatively and postoperatively included evaluation of graft patency, hind limb function, and biochemical markers of tissue ischemia or reperfusion injury. Histological analysis was performed on explants to assess host cell responses. RESULTS Postoperative gait assessment and biochemical analysis confirmed that ischemia and reperfusion injury were caused by 6-hour ischemia, regardless of vascular graft type. Hind limb function and tissue damage biomarkers improved in all groups postoperatively. Final patency rates at postoperative day 28 were higher for HAV than for ePTFE graft in both the 0-hour (HAV, 85.7%; ePTFE, 66.7%) and 6-hour (HAV, 100%; ePTFE, 75%) ischemia groups, but these differences were not statistically significant. Histological analyses identified some intimal hyperplasia and host reactivity to the xenogeneic HAV and also to the synthetic ePTFE graft. Positive host integration and vascular cell infiltration were identified in HAV but not ePTFE explants. CONCLUSION Based on the functional performance and the histologic profile of explanted HAVs, this study supports further investigation to evaluate long-term performance of the HAV when used to repair traumatic vascular injuries.
Collapse
|
35
|
Yan J, Xiao H, Zhou X, Li Y, Zhao S, Zhao X, Liu Y, Liu M, Xue F, Zhang Q, Zhao W, Li L, Su Y, Zeng W. Engineered exosomes reprogram Gli1 + cells in vivo to prevent calcification of vascular grafts and autologous pathological vessels. SCIENCE ADVANCES 2023; 9:eadf7858. [PMID: 37478186 PMCID: PMC10361604 DOI: 10.1126/sciadv.adf7858] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 06/20/2023] [Indexed: 07/23/2023]
Abstract
Calcification of autologous pathological vessels and tissue engineering blood vessels (TEBVs) is a thorny problem in clinic. However, there is no effective and noninvasive treatment that is available against the calcification of TEBVs and autologous pathological vessels. Gli1+ cells are progenitors of smooth muscle cells (SMCs) and can differentiate into osteoblast-like cells, leading to vascular calcification. Our results showed that the spatiotemporal distribution of Gli1+ cells in TEBVs was positively correlated with the degree of TEBV calcification. An anticalcification approach was designed consisting of exosomes derived from mesenchymal stem cells delivering lncRNA-ANCR to construct the engineered exosome-Ancr/E7-EXO. The results showed that Ancr/E7-EXO effectively targeted Gli1+ cells, promoting rapid SMC reconstruction and markedly inhibiting Gli1+ cell differentiation into osteoblast-like cells. Moreover, Ancr/E7-EXO significantly inhibited vascular calcification caused by chronic kidney disease. Therefore, Ancr/E7-EXO reprogrammed Gli1+ cells to prevent calcification of vascular graft and autologous pathological vessel, providing unique insights for an effective anticalcification.
Collapse
Affiliation(s)
- Juan Yan
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Haoran Xiao
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Xin Zhou
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Yanzhao Li
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing 400038, China
| | - Shanlan Zhao
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Xingli Zhao
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Yong Liu
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State and Local Joint Engineering Laboratory for Vascular Implants, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing 400038, China
| | - Min Liu
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Fangchao Xue
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Qiao Zhang
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Wenyan Zhao
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Lang Li
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Yang Su
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Army Medical University, Chongqing 400038, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
36
|
Brown TK, Alharbi S, Ho KJ, Jiang B. Prosthetic vascular grafts engineered to combat calcification: Progress and future directions. Biotechnol Bioeng 2023; 120:953-969. [PMID: 36544433 PMCID: PMC10023339 DOI: 10.1002/bit.28316] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Calcification in prosthetic vascular conduits is a major challenge in cardiac and vascular surgery that compromises the long-term performance of these devices. Significant research efforts have been made to understand the etiology of calcification in the cardiovascular system and to combat calcification in various cardiovascular devices. Novel biomaterial design and tissue engineering strategies have shown promise in preventing or delaying calcification in prosthetic vascular grafts. In this review, we highlight recent advancements in the development of acellular prosthetic vascular grafts with preclinical success in attenuating calcification through advanced biomaterial design. We also discuss the mechanisms of action involved in the designs that will contribute to the further understanding of cardiovascular calcification. Lastly, recent insights into the etiology of vascular calcification will guide the design of future prosthetic vascular grafts with greater potential for translational success.
Collapse
Affiliation(s)
- Taylor K. Brown
- Department of Biomedical Engineering, Northwestern University, Chicago, IL
| | - Sara Alharbi
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Karen J. Ho
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Bin Jiang
- Department of Biomedical Engineering, Northwestern University, Chicago, IL
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
37
|
Barungi S, Hernández-Camarero P, Moreno-Terribas G, Villalba-Montoro R, Marchal JA, López-Ruiz E, Perán M. Clinical implications of inflammation in atheroma formation and novel therapies in cardiovascular diseases. Front Cell Dev Biol 2023; 11:1148768. [PMID: 37009489 PMCID: PMC10061140 DOI: 10.3389/fcell.2023.1148768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Cardiovascular diseases (CVD) are the leading causes of death and disability in the world. Among all CVD, the most common is coronary artery disease (CAD). CAD results from the complications promoted by atherosclerosis, which is characterized by the accumulation of atherosclerotic plaques that limit and block the blood flow of the arteries involved in heart oxygenation. Atherosclerotic disease is usually treated by stents implantation and angioplasty, but these surgical interventions also favour thrombosis and restenosis which often lead to device failure. Hence, efficient and long-lasting therapeutic options that are easily accessible to patients are in high demand. Advanced technologies including nanotechnology or vascular tissue engineering may provide promising solutions for CVD. Moreover, advances in the understanding of the biological processes underlying atherosclerosis can lead to a significant improvement in the management of CVD and even to the development of novel efficient drugs. To note, over the last years, the observation that inflammation leads to atherosclerosis has gained interest providing a link between atheroma formation and oncogenesis. Here, we have focused on the description of the available therapy for atherosclerosis, including surgical treatment and experimental treatment, the mechanisms of atheroma formation, and possible novel therapeutic candidates such as the use of anti-inflammatory treatments to reduce CVD.
Collapse
Affiliation(s)
- Shivan Barungi
- Department of Health Sciences, University of Jaén, Jaén, Spain
| | | | | | | | - Juan Antonio Marchal
- Centre for Biomedical Research (CIBM), Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, Granada, Spain
| | - Elena López-Ruiz
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Centre for Biomedical Research (CIBM), Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, Granada, Spain
| |
Collapse
|
38
|
Ratner B. Vascular Grafts: Technology Success/Technology Failure. BME FRONTIERS 2023; 4:0003. [PMID: 37849668 PMCID: PMC10521696 DOI: 10.34133/bmef.0003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/15/2022] [Indexed: 10/19/2023] Open
Abstract
Vascular prostheses (grafts) are widely used for hemodialysis blood access, trauma repair, aneurism repair, and cardiovascular reconstruction. However, smaller-diameter (≤4 mm) grafts that would be valuable for many reconstructions have not been achieved to date, although hundreds of papers on small-diameter vascular grafts have been published. This perspective article presents a hypothesis that may open new research avenues for the development of small-diameter vascular grafts. A historical review of the vascular graft literature and specific types of vascular grafts is presented focusing on observations important to the hypothesis to be presented. Considerations in critically reviewing the vascular graft literature are discussed. The hypothesis that perhaps the "biocompatible biomaterials" comprising our vascular grafts-biomaterials that generate dense, nonvascularized collagenous capsules upon implantation-may not be all that biocompatible is presented. Examples of materials that heal with tissue reconstruction and vascularity, in contrast to the fibrotic encapsulation, are offered. Such prohealing materials may lead the way to a new generation of vascular grafts suitable for small-diameter reconstructions.
Collapse
Affiliation(s)
- Buddy Ratner
- Center for Dialysis Innovation (CDI), Departments of Bioengineering and Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
39
|
Shakeel A, Corridon PR. Mitigating challenges and expanding the future of vascular tissue engineering-are we there yet? Front Physiol 2023; 13:1079421. [PMID: 36685187 PMCID: PMC9846051 DOI: 10.3389/fphys.2022.1079421] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Adeeba Shakeel
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter R. Corridon
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biomedical Engineering, Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
40
|
Gutowski P, Guziewicz M, Ilzecki M, Kazimierczak A, Lawson JH, Prichard HL, Przywara S, Samad R, Tente W, Turek J, Witkiewicz W, Zapotoczny N, Zubilewicz T, Niklason LE. Six-year outcomes of a phase II study of human-tissue engineered blood vessels for peripheral arterial bypass. JVS Vasc Sci 2023; 4:100092. [PMID: 36874956 PMCID: PMC9976461 DOI: 10.1016/j.jvssci.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Objective The human acellular vessel (HAV) was evaluated for surgical bypass in a phase II study. The primary results at 24 months after implantation have been reported, and the patients will be evaluated for ≤10 years. Methods In the present report, we have described the 6-year results of a prospective, open-label, single-treatment arm, multicenter study. Patients with advanced peripheral artery disease (PAD) requiring above-the-knee femoropopliteal bypass surgery without available autologous graft options had undergone implantation with the HAV, a bioengineered human tissue replacement blood vessel. The patients who completed the 24-month primary portion of the study will be evaluated for ≤10 years after implantation. The present mid-term analysis was performed at the 6-year milestone (72 months) for patients followed up for 24 to 72 months. Results HAVs were implanted in 20 patients at three sites in Poland. Seven patients had discontinued the study before completing the 2-year portion of the study: four after graft occlusion had occurred and three who had died of causes deemed unrelated to the conduit, with the HAV reported as functional at their last visit. The primary results at 24 months showed primary, primary assisted, and secondary patency rates of 58%, 58%, and 74%, respectively. One vessel had developed a pseudoaneurysm deemed possibly iatrogenic; no other signs of structural failure were reported. No rejections or infections of the HAV occurred, and no patient had required amputation of the implanted limb. Of the 20 patients, 13 had completed the primary portion of the study; however, 1 patient had died shortly after 24 months. Of the remaining 12 patients, 3 died of causes unrelated to the HAV. One patient had required thrombectomy twice, with secondary patency achieved. No other interventions were recorded between 24 and 72 months. At 72 months, five patients had a patent HAV, including four patients with primary patency. For the entire study population from day 1 to month 72, the overall primary, primary assisted, and secondary patency rate estimated using Kaplan-Meier analysis was 44%, 45%, and 60% respectively, with censoring for death. No patient had experienced rejection or infection of the HAV, and no patient had required amputation of the implanted limb. Conclusions The infection-resistant, off-the-shelf HAV could provide a durable alternative conduit in the arterial circuit setting to restore the lower extremity blood supply in patients with PAD, with remodeling into the recipient's own vessel over time. The HAV is currently being evaluated in seven clinical trials to treat PAD, vascular trauma, and as a hemodialysis access conduit.
Collapse
Affiliation(s)
- Piotr Gutowski
- Department of Vascular Surgery and Angiology, Pomeranian Medical University of Szczecin, Szczecin, Poland
| | - Malgorzata Guziewicz
- Research and Development Centre, Department of Vascular Surgery, General Hospital, Wroclaw, Poland
| | - Marek Ilzecki
- Clinic of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Arkadiusz Kazimierczak
- Department of Vascular Surgery and Angiology, Pomeranian Medical University of Szczecin, Szczecin, Poland
| | - Jeffrey H. Lawson
- Humacyte, Inc, Durham, NC
- Department of Surgery, Duke University, Durham, NC
| | | | - Stanislaw Przywara
- Clinic of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Rabih Samad
- Department of Vascular Surgery and Angiology, Pomeranian Medical University of Szczecin, Szczecin, Poland
| | | | - Jakub Turek
- Research and Development Centre, Department of Vascular Surgery, General Hospital, Wroclaw, Poland
| | - Wojcieh Witkiewicz
- Research and Development Centre, Department of Vascular Surgery, General Hospital, Wroclaw, Poland
| | - Norbert Zapotoczny
- Research and Development Centre, Department of Vascular Surgery, General Hospital, Wroclaw, Poland
| | - Tomaz Zubilewicz
- Clinic of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Laura E. Niklason
- Humacyte, Inc, Durham, NC
- Department of Anesthesia and Biomedical Engineering, Yale University, New Haven
- Department of Biomedical Engineering, Yale University, New Haven, CT
- Correspondence: Laura E. Niklason, MD, PhD, Humacyte, Inc, 2525 NC-54, Durham, NC 27713
| |
Collapse
|
41
|
Antonova L, Kutikhin A, Sevostianova V, Lobov A, Repkin E, Krivkina E, Velikanova E, Mironov A, Mukhamadiyarov R, Senokosova E, Khanova M, Shishkova D, Markova V, Barbarash L. Controlled and Synchronised Vascular Regeneration upon the Implantation of Iloprost- and Cationic Amphiphilic Drugs-Conjugated Tissue-Engineered Vascular Grafts into the Ovine Carotid Artery: A Proteomics-Empowered Study. Polymers (Basel) 2022; 14:polym14235149. [PMID: 36501545 PMCID: PMC9736446 DOI: 10.3390/polym14235149] [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: 10/23/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Implementation of small-diameter tissue-engineered vascular grafts (TEVGs) into clinical practice is still delayed due to the frequent complications, including thrombosis, aneurysms, neointimal hyperplasia, calcification, atherosclerosis, and infection. Here, we conjugated a vasodilator/platelet inhibitor, iloprost, and an antimicrobial cationic amphiphilic drug, 1,5-bis-(4-tetradecyl-1,4-diazoniabicyclo [2.2.2]octan-1-yl) pentane tetrabromide, to the luminal surface of electrospun poly(ε-caprolactone) (PCL) TEVGs for preventing thrombosis and infection, additionally enveloped such TEVGs into the PCL sheath to preclude aneurysms, and implanted PCLIlo/CAD TEVGs into the ovine carotid artery (n = 12) for 6 months. The primary patency was 50% (6/12 animals). TEVGs were completely replaced with the vascular tissue, free from aneurysms, calcification, atherosclerosis and infection, completely endothelialised, and had clearly distinguishable medial and adventitial layers. Comparative proteomic profiling of TEVGs and contralateral carotid arteries found that TEVGs lacked contractile vascular smooth muscle cell markers, basement membrane components, and proteins mediating antioxidant defense, concurrently showing the protein signatures of upregulated protein synthesis, folding and assembly, enhanced energy metabolism, and macrophage-driven inflammation. Collectively, these results suggested a synchronised replacement of PCL with a newly formed vascular tissue but insufficient compliance of PCLIlo/CAD TEVGs, demanding their testing in the muscular artery position or stimulation of vascular smooth muscle cell specification after the implantation.
Collapse
Affiliation(s)
- Larisa Antonova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
- Correspondence: ; Tel.: +7-9609077067
| | - Viktoriia Sevostianova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Arseniy Lobov
- Department of Regenerative Biomedicine, Research Institute of Cytology, 4 Tikhoretskiy Prospekt, Saint Petersburg 194064, Russia
| | - Egor Repkin
- Centre for Molecular and Cell Technologies, Saint Petersburg State University, Universitetskaya Embankment, 7/9, Saint Petersburg 199034, Russia
| | - Evgenia Krivkina
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Elena Velikanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Andrey Mironov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Rinat Mukhamadiyarov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Evgenia Senokosova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Mariam Khanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Leonid Barbarash
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| |
Collapse
|
42
|
Rizzi S, Mantero S, Boschetti F, Pesce M. Luminal endothelialization of small caliber silk tubular graft for vascular constructs engineering. Front Cardiovasc Med 2022; 9:1013183. [PMID: 36465472 PMCID: PMC9708712 DOI: 10.3389/fcvm.2022.1013183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/31/2022] [Indexed: 09/29/2023] Open
Abstract
The constantly increasing incidence of coronary artery disease worldwide makes necessary to set advanced therapies and tools such as tissue engineered vessel grafts (TEVGs) to surpass the autologous grafts [(i.e., mammary and internal thoracic arteries, saphenous vein (SV)] currently employed in coronary artery and vascular surgery. To this aim, in vitro cellularization of artificial tubular scaffolds still holds a good potential to overcome the unresolved problem of vessel conduits availability and the issues resulting from thrombosis, intima hyperplasia and matrix remodeling, occurring in autologous grafts especially with small caliber (<6 mm). The employment of silk-based tubular scaffolds has been proposed as a promising approach to engineer small caliber cellularized vascular constructs. The advantage of the silk material is the excellent manufacturability and the easiness of fiber deposition, mechanical properties, low immunogenicity and the extremely high in vivo biocompatibility. In the present work, we propose a method to optimize coverage of the luminal surface of silk electrospun tubular scaffold with endothelial cells. Our strategy is based on seeding endothelial cells (ECs) on the luminal surface of the scaffolds using a low-speed rolling. We show that this procedure allows the formation of a nearly complete EC monolayer suitable for flow-dependent studies and vascular maturation, as a step toward derivation of complete vascular constructs for transplantation and disease modeling.
Collapse
Affiliation(s)
- Stefano Rizzi
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Ph.D. Program in Biomedical Engineering, Politecnico di Milano, Milan, Italy
| | - Sara Mantero
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Federica Boschetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | | |
Collapse
|
43
|
Engineering Smooth Muscle to Understand Extracellular Matrix Remodeling and Vascular Disease. Bioengineering (Basel) 2022; 9:bioengineering9090449. [PMID: 36134994 PMCID: PMC9495899 DOI: 10.3390/bioengineering9090449] [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: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The vascular smooth muscle is vital for regulating blood pressure and maintaining cardiovascular health, and the resident smooth muscle cells (SMCs) in blood vessel walls rely on specific mechanical and biochemical signals to carry out these functions. Any slight change in their surrounding environment causes swift changes in their phenotype and secretory profile, leading to changes in the structure and functionality of vessel walls that cause pathological conditions. To adequately treat vascular diseases, it is essential to understand how SMCs crosstalk with their surrounding extracellular matrix (ECM). Here, we summarize in vivo and traditional in vitro studies of pathological vessel wall remodeling due to the SMC phenotype and, conversely, the SMC behavior in response to key ECM properties. We then analyze how three-dimensional tissue engineering approaches provide opportunities to model SMCs’ response to specific stimuli in the human body. Additionally, we review how applying biomechanical forces and biochemical stimulation, such as pulsatile fluid flow and secreted factors from other cell types, allows us to study disease mechanisms. Overall, we propose that in vitro tissue engineering of human vascular smooth muscle can facilitate a better understanding of relevant cardiovascular diseases using high throughput experiments, thus potentially leading to therapeutics or treatments to be tested in the future.
Collapse
|