1
|
Miao X, Chen T, Lang Z, Wu Y, Wu X, Zhu Z, Xu RX. Design, fabrication, and application of bioengineering vascular networks based on microfluidic strategies. J Mater Chem B 2025; 13:1252-1269. [PMID: 39691980 DOI: 10.1039/d4tb02047b] [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: 12/19/2024]
Abstract
Vascularization is a critical component of tissue engineering research and is essential for enhancing the success rate of tissue construction and function. Over the past decade, researchers have explored various methods to construct in vitro vascular networks, including 3D printing, cell sphere technology, and microfluidics. Microfluidic technology has garnered significant attention due to its notable advantages in precision, controllability, flexibility, and applicability. It can be primarily classified into two modes: (i) the pre-designed mode, which involves creating vascular networks by pre-designing vascular channels and seeding endothelial cells, encompassing microfluidic chips and microfluidic spinning technologies; and (ii) the self-assembly mode, where cell spheres are fabricated using microfluidic technology and subsequently self-assemble into vascular networks. In this review, we first provide a brief overview of the normal physiological and pathological characteristics of vascular networks, followed by a discussion of the factors to be considered in designing in vitro vascular networks, and conclude with an examination of the classification of technologies for the preparation of microfluidic vascular networks and recent advancements. It is anticipated that in vitro vascular network models will soon be successfully applied in regenerative medicine and drug development.
Collapse
Affiliation(s)
- Xiaoping Miao
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Tianao Chen
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhongliang Lang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
- Department of Plastic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China.
| | - Yongqi Wu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Xizhi Wu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhiqiang Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China.
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Ronald X Xu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, 215123, P. R. China
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
2
|
Chatzopoulou E, Bousaidi N, Guilbert T, Rucher G, Rose J, Germain S, Rouzet F, Chaussain C, Muller L, Gorin C. Multiscale Imaging to Monitor Functional SHED-Supported Engineered Vessels. J Dent Res 2024; 103:1392-1402. [PMID: 39290146 DOI: 10.1177/00220345241271122] [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: 09/19/2024] Open
Abstract
Regeneration of orofacial tissues is hampered by the lack of adequate vascular supply. Implantation of in vitro engineered, prevascularized constructs has emerged as a strategy to allow the rapid vascularization of the entire graft. Given the angiogenic properties of dental pulp stem cells, we hereby established a preclinical model of prevascularized constructs loaded with stem cells from human exfoliating deciduous teeth (SHED) in a 3-dimensional-printed material and provided a functional analysis of their in vivo angiogenesis, vascular perfusion, and permeability. Three different cell-loaded collagen hydrogels (SHED-human umbilical vein endothelial cell [HUVEC], HUVEC with SHED-conditioned medium, and SHED alone) were cast in polylactic acid (PLA) grids and ectopically implanted in athymic mice. At day 10, in vivo positron emission tomography (PETscan) revealed a significantly increased uptake of radiotracer targeting activated endothelial cells in the SHED-HUVEC group compared to the other groups. At day 30, ex vivo micro-computed tomography imaging confirmed that SHED-HUVEC constructs had a significantly increased vascular volume compared to the other ones. Injection of species-specific lectins analyzed by 2-photon microscopy demonstrated blood perfusion of the engineered human vessels in both prevascularized groups. However, in vivo quantification showed increased vessel density in the SHED-HUVEC group. In addition, coinjection of fluorescent lectin and dextran revealed that prevascularization with SHED prevented vascular leakage, demonstrating the active role of SHED in the maturation of human-engineered microvascular networks. This preclinical study introduces a novel PLA prevascularized and implantable construct, along with an array of imaging techniques, to validate the ability of SHED to promote functional human-engineered vessels, further highlighting the interest of SHED for orofacial tissue engineering. Furthermore, this study validates the use of PETscan for the early detection of in vivo angiogenesis, which may be applied in the clinic to monitor the performance of prevascularized grafts.
Collapse
Affiliation(s)
- E Chatzopoulou
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| | - N Bousaidi
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
| | - T Guilbert
- Université Paris Cité, CNRS, INSERM U1016, Institut Cochin, Paris, France
| | - G Rucher
- Université Paris Cité, LVTS, INSERM U1148, France
- Université Paris Cité, UMS 34-FRIM, France
| | - J Rose
- AP-HP, Département de médecine nucléaire, Hôpital Bichat, Paris, France
| | - S Germain
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, Université PSL, Paris, France
| | - F Rouzet
- Université Paris Cité, LVTS, INSERM U1148, France
- AP-HP, Département de médecine nucléaire, Hôpital Bichat, Paris, France
| | - C Chaussain
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| | - L Muller
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, Université PSL, Paris, France
| | - C Gorin
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| |
Collapse
|
3
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024; 27:587-621. [PMID: 38842751 PMCID: PMC11564345 DOI: 10.1007/s10456-024-09928-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Ling Z, Zhang H, Zhao J, Wang P, An Z, Xiao S, Sun Y, Fu W. Electrostimulation-Based Decellularized Matrix Bladder Patch Promotes Bladder Repair in Rats. ACS Biomater Sci Eng 2024; 10:6498-6508. [PMID: 39240226 DOI: 10.1021/acsbiomaterials.4c00961] [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: 09/07/2024]
Abstract
Bladder tissue engineering offers significant potential for repairing defects resulting from congenital and acquired conditions. However, the effectiveness of engineered grafts is often constrained by insufficient vascularization and neural regeneration. This study utilized four primary biomaterials─gelatin methacryloyl (GelMA), chitin nanocrystals (ChiNC), titanium carbide (MXene), and adipose-derived stem cells (ADSC)─to formulate two types of bioinks, GCM0.2 and GCM0.2-ADSC, in specified proportions. These bioinks were 3D printed onto bladder acellular matrix (BAM) patches to create BAM-GCM0.2 and BAM-GCM0.2-ADSC patches. The BAM-GCM0.2-ADSC patches underwent electrical stimulation to yield GCM0.2-ADSC-ES bladder patches. Employed for the repair of rat bladder defects, these patches were evaluated against a Control group, which underwent partial cystectomy followed by direct suturing. Our findings indicate that the inclusion of ADSC and electrical stimulation significantly enhances the regeneration of rat bladder smooth muscle (from [24.052 ± 2.782] % to [57.380 ± 4.017] %), blood vessels (from [5.326 ± 0.703] % to [12.723 ± 1.440] %), and nerves (from [0.227 ± 0.017] % to [1.369 ± 0.218] %). This research underscores the superior bladder repair capabilities of the GCM0.2-ADSC-ES patch and opens new pathways for bladder defect repair.
Collapse
Affiliation(s)
- Zhengyun Ling
- Department of Urology, The Third Medical Center, PLA General Hospital, Beijing 100039, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoqian Zhang
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Jian Zhao
- Department of Urology, 960th Hospital of PLA, Jinan 250031, China
| | | | - Ziyan An
- Medical School of PLA, Beijing 100853, China
| | - Shuwei Xiao
- Department of Urology, Air Force Medical Center, Beijing 100142, China
| | - Yanfeng Sun
- Department of Pediatrics, The Third Medical Center, PLA General Hospital, Beijing 100039, China
| | - Weijun Fu
- Department of Urology, The Third Medical Center, PLA General Hospital, Beijing 100039, China
| |
Collapse
|
5
|
Wang Y, Liu M, Zhang W, Liu H, Jin F, Mao S, Han C, Wang X. Mechanical strategies to promote vascularization for tissue engineering and regenerative medicine. BURNS & TRAUMA 2024; 12:tkae039. [PMID: 39350780 PMCID: PMC11441985 DOI: 10.1093/burnst/tkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 10/04/2024]
Abstract
Vascularization is a major challenge in the field of tissue engineering and regenerative medicine. Mechanical factors have been demonstrated to play a fundamental role in vasculogenesis and angiogenesis and can affect the architecture of the generated vascular network. Through the regulation of mechanical factors in engineered tissues, various mechanical strategies can be used to optimize the preformed vascular network and promote its rapid integration with host vessels. Optimization of the mechanical properties of scaffolds, including controlling scaffold stiffness, increasing surface roughness and anisotropic structure, and designing interconnected, hierarchical pore structures, is beneficial for the in vitro formation of vascular networks and the ingrowth of host blood vessels. The incorporation of hollow channels into scaffolds promotes the formation of patterned vascular networks. Dynamic stretching and perfusion can facilitate the formation and maturation of preformed vascular networks in vitro. Several indirect mechanical strategies provide sustained mechanical stimulation to engineered tissues in vivo, which further promotes the vascularization of implants within the body. Additionally, stiffness gradients, anisotropic substrates and hollow channels in scaffolds, as well as external cyclic stretch, boundary constraints and dynamic flow culture, can effectively regulate the alignment of vascular networks, thereby promoting better integration of prevascularized engineered tissues with host blood vessels. This review summarizes the influence and contribution of both scaffold-based and external stimulus-based mechanical strategies for vascularization in tissue engineering and elucidates the underlying mechanisms involved.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Meixuan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Huan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Fang Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Shulei Mao
- Department of Burns and Plastic Surgery, Quhua Hospital of Zhejiang, 62 Wenchang Road, Quhua, Quzhou 324004, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| |
Collapse
|
6
|
Dinter MC, Bickelmann C, Nickels RM, Menger MD, Laschke MW. Microvascular Fragment-Loaded Platelet-Rich Plasma Dressing Promotes Cutaneous Wound Healing. Adv Wound Care (New Rochelle) 2024; 13:336-349. [PMID: 38299944 DOI: 10.1089/wound.2023.0029] [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: 02/02/2024] Open
Abstract
Objective: Chronic wounds represent a considerable burden for the affected patients and the health care system. To overcome this problem, effective treatment strategies are urgently required. In this study, we tested a novel approach by combining platelet-rich plasma (PRP) and microvascular fragments (MVF) to create a prevascularized gel dressing. Approach: MVF were enzymatically isolated from the epididymal fat pads of transgenic green fluorescent protein (GFP)+ C57BL/6J donor mice. Subsequently, 5,000 MVF were suspended in 10 μL murine PRP as carrier and transferred into full-thickness skin wounds within dorsal skinfold chambers of C57BL/6J wild-type mice (PRP+MVF). Wound healing in comparison to empty wounds (control) and wounds filled with PRP alone was repeatedly analyzed throughout 14 days by means of stereomicroscopy, histology, and immunohistochemistry. Results: Planimetric assessment of the wound size over time revealed a significantly accelerated and improved healing of PRP+MVF-treated wounds when compared with PRP-treated and empty control wounds. These wounds also exhibited a significantly higher density of blood and lymph vessels, which originated from the GFP+ MVF isolates and effectively promoted granulation tissue formation inside the skin defects. Innovation: This study is the first to combine PRP and MVF for the improvement of wound healing. Conclusion: The combination of PRP and MVF represents a promising approach for the future treatment of wounds that do not heal spontaneously due to poor wound-healing conditions.
Collapse
Affiliation(s)
- Melina C Dinter
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Caroline Bickelmann
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Ruth M Nickels
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
7
|
Chandra Sekar N, Khoshmanesh K, Baratchi S. Bioengineered models of cardiovascular diseases. Atherosclerosis 2024; 393:117565. [PMID: 38714426 DOI: 10.1016/j.atherosclerosis.2024.117565] [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: 11/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024]
Abstract
Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.
Collapse
Affiliation(s)
- Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
8
|
Mayer SA, Thomas B, Heuer M, Brune JC, Eras V, Schuster K, Knoedler L, Schaefer RL, Thiele W, Sleeman JP, Dimmler A, Heimel P, Kneser U, Bigdeli AK, Falkner F. In Vivo Engineering and Transplantation of Axially Vascularized and Epithelialized Flaps in Rats. Tissue Eng Part A 2024. [PMID: 38623816 DOI: 10.1089/ten.tea.2024.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
The arteriovenous loop (AVL) model allows the in vivo engineering of axially vascularized flaps, the so-called AVL flaps. Although AVL flaps can be transplanted microsurgically to cover tissue defects, they lack an epithelial layer on the surface. Therefore, the objective of this study was to engineer axially vascularized AVL flaps with an accompanying epithelial layer for local defect reconstruction. In this study, AVLs were established in 20 male Lewis rats. Minimally invasive injection of keratinocytes onto the surface of the AVL flaps was performed on postoperative day (POD) 21. AVL flaps were explanted from 12 rats on POD 24 or POD 30, then the epithelium formed by the keratinocytes on the surface of the flaps was evaluated using immunofluorescence staining. In six other rats, the AVL flap was locally transposed to cover a critical defect in the rats' leg on POD 30 and explanted for analysis on POD 40. In two control rats, sodium chloride was applied instead of keratinocytes. These control flaps were also transplanted on POD 30 and explanted on POD 40. Our results revealed that 3 days after keratinocyte application, a loose single-layered epithelium was observed histologically on the AVL flaps surface, whereas after 9 days, a multilayered and structured epithelium had grown. The epithelium on the transplanted AVL flaps showed its physiological differentiation when being exposed to an air-liquid interface. Histologically, a layered epithelium identical to the rats' regular skin was formed. In the sodium chloride control group, no epithelium had been grown. This study clearly demonstrates that axially vascularized AVL flaps can be processed in the subcutaneous chamber by minimally invasive injection of keratinocytes. Thus, AVL flaps with an intact epithelial layer were engineered and could be successfully transplanted for local defect coverage in a small animal model.
Collapse
Affiliation(s)
- Simon Andreas Mayer
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
- Department of Hand, Plastic, and Reconstructive Surgery, University of Heidelberg, Heidelberg, Germany
| | - Benjamin Thomas
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
- Department of Hand, Plastic, and Reconstructive Surgery, University of Heidelberg, Heidelberg, Germany
| | - Miriam Heuer
- German Institute for Cell and Tissue Replacement, Berlin, Germany
| | - Jan C Brune
- German Institute for Cell and Tissue Replacement, Berlin, Germany
| | - Volker Eras
- German Institute for Cell and Tissue Replacement, Berlin, Germany
| | - Kilian Schuster
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
- Department of Hand, Plastic, and Reconstructive Surgery, University of Heidelberg, Heidelberg, Germany
| | - Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Luisa Schaefer
- Department of Hand, Plastic, and Reconstructive Surgery, University of Freiburg, Freiburg, Germany
| | - Wilko Thiele
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jonathan P Sleeman
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute for Biological and Chemical Systems, Karlsruhe Institute of Technology (KIT), Campus North, Karlsruhe, Germany
| | - Arno Dimmler
- Institute of Pathology, Vincentius Kliniken Karlsruhe, Karlsruhe, Germany
| | - Patrick Heimel
- Core Facility Hard Tissue and Biomaterial Research, Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology the Research Center in Cooperation with AUVA, Vienna, Austria
| | - Ulrich Kneser
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
- Department of Hand, Plastic, and Reconstructive Surgery, University of Heidelberg, Heidelberg, Germany
| | - Amir K Bigdeli
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
- Department of Hand, Plastic, and Reconstructive Surgery, University of Heidelberg, Heidelberg, Germany
| | - Florian Falkner
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
- Department of Hand, Plastic, and Reconstructive Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Song H, Hao D, Zhou J, Farmer D, Wang A. Development of pro-angiogenic skin substitutes for wound healing. Wound Repair Regen 2024; 32:208-216. [PMID: 38308588 DOI: 10.1111/wrr.13154] [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: 08/21/2023] [Revised: 11/13/2023] [Accepted: 12/12/2023] [Indexed: 02/05/2024]
Abstract
Wounds pose significant challenges to public health, primarily due to the loss of the mechanical integrity and barrier function of the skin and impaired angiogenesis, causing physical morbidities and psychological trauma to affect patients. Reconstructing the vasculature of the wound bed is crucial for promoting wound healing, reducing scar formation and enhancing the quality of life for patients. The development of pro-angiogenic skin substitutes has emerged as a promising strategy to facilitate vascularization and expedite the healing process of burn wounds. This review provides an overview of the various types of skin substitutes employed in wound healing, explicitly emphasising those designed to enhance angiogenesis. Synthetic scaffolds, biological matrices and tissue-engineered constructs incorporating stem cells and primary cells, cell-derived extracellular vesicles (EVs), pro-angiogenic growth factors and peptides, as well as gene therapy-based skin substitutes are thoroughly examined. The review summarises the existing challenges, future directions and potential innovations in pro-angiogenic dressing for skin substitutes. It highlights the need for continued research to develop new technologies and combine multiple strategies and factors, and to overcome obstacles and advance the field, ultimately leading to improved outcomes for wound patients.
Collapse
Affiliation(s)
- Hengyue Song
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Dake Hao
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Diana Farmer
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
- Department of Biomedical Engineering, UC Davis, Davis, California, USA
| |
Collapse
|
10
|
Mazio C, Mavaro I, Palladino A, Casale C, Urciuolo F, Banfi A, D'Angelo L, Netti PA, de Girolamo P, Imparato G, Attanasio C. Rapid innervation and physiological epidermal regeneration by bioengineered dermis implanted in mouse. Mater Today Bio 2024; 25:100949. [PMID: 38298559 PMCID: PMC10827562 DOI: 10.1016/j.mtbio.2024.100949] [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: 09/25/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 02/02/2024] Open
Abstract
Tissue-engineered skin substitutes are promising tools to cover large and deep skin defects. However, the lack of a synergic and fast regeneration of the vascular network, nerves, and skin appendages limits complete skin healing and impairs functional recovery. It has been highlighted that an ideal skin substitute should mimic the structure of the native tissue to enhance clinical effectiveness. Here, we produced a pre-vascularized dermis (PVD) comprised of fibroblasts embedded in their own extracellular matrix (ECM) and a capillary-like network. Upon implantation in a mouse full-thickness skin defect model, we observed a very early innervation of the graft in 2 weeks. In addition, mouse capillaries and complete epithelialization were detectable as early as 1 week after implantation and, skin appendages developed in 2 weeks. These anatomical features underlie the interaction with the skin nerves, thus providing a further cue for reinnervation guidance. Further, the graft displays mechanical properties, collagen density, and assembly features very similar to the host tissue. Taken together our data show that the pre-existing ECM components of the PVD, physiologically organized and assembled similarly to the native tissue, support a rapid regeneration of dermal tissue. Therefore, our results suggest a promising potential for PVD in skin regeneration.
Collapse
Affiliation(s)
- Claudia Mazio
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
| | - Isabella Mavaro
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| | - Antonio Palladino
- University of Naples Federico II, Department of Agricultural Sciences, Italy
| | - Costantino Casale
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Italy
| | - Francesco Urciuolo
- University of Naples Federico II, Department of Chemical, Materials and Industrial Production Engineering, Italy
| | - Andrea Banfi
- Basel University Hospital and University of Basel, Department of Biomedicine, Switzerland
| | - Livia D'Angelo
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| | - Paolo A. Netti
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Italy
- University of Naples Federico II, Department of Chemical, Materials and Industrial Production Engineering, Italy
| | - Paolo de Girolamo
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| | - Giorgia Imparato
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
| | - Chiara Attanasio
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| |
Collapse
|
11
|
Friend NE, Beamish JA, Margolis EA, Schott NG, Stegemann JP, Putnam AJ. Pre-cultured, cell-encapsulating fibrin microbeads for the vascularization of ischemic tissues. J Biomed Mater Res A 2024; 112:549-561. [PMID: 37326361 PMCID: PMC10724379 DOI: 10.1002/jbm.a.37580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
There is a significant clinical need to develop effective vascularization strategies for tissue engineering and the treatment of ischemic pathologies. In patients afflicted with critical limb ischemia, comorbidities may limit common revascularization strategies. Cell-encapsulating modular microbeads possess a variety of advantageous properties, including the ability to support prevascularization in vitro while retaining the ability to be injected in a minimally invasive manner in vivo. Here, fibrin microbeads containing human umbilical vein endothelial cells (HUVEC) and bone marrow-derived mesenchymal stromal cells (MSC) were cultured in suspension for 3 days (D3 PC microbeads) before being implanted within intramuscular pockets in a SCID mouse model of hindlimb ischemia. By 14 days post-surgery, animals treated with D3 PC microbeads showed increased macroscopic reperfusion of ischemic foot pads and improved limb salvage compared to the cellular controls. Delivery of HUVEC and MSC via microbeads led to the formation of extensive microvascular networks throughout the implants. Engineered vessels of human origins showed evidence of inosculation with host vasculature, as indicated by erythrocytes present in hCD31+ vessels. Over time, the total number of human-derived vessels within the implant region decreased as networks remodeled and an increase in mature, pericyte-supported vascular structures was observed. Our findings highlight the potential therapeutic benefit of developing modular, prevascularized microbeads as a minimally invasive therapeutic for treating ischemic tissues.
Collapse
Affiliation(s)
- Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Jeffrey A. Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Emily A. Margolis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | | | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| |
Collapse
|
12
|
Wei P, Wu L, Xie H, Chen Z, Tan R, Xu Z. Application of a meshed artificial dermal scaffold and negative-pressure wound therapy in the treatment of full-thickness skin defects: a prospective in vivo study. Biomater Sci 2024; 12:1914-1923. [PMID: 38436071 DOI: 10.1039/d3bm01675g] [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: 03/05/2024]
Abstract
Artificial dermal scaffolds (ADSs) have great value in repairing deep skin defects. However, problems such as unsatisfactory angiogenesis and local dropsy or empyema often occur, resulting in delayed or even failed wound healing. Negative pressure wound therapy (NPWT) is an effective therapy to promote wound healing or shorten wound bed preparation time. Studies on whether it can improve the effects of ADSs have never been interrupted, and no consensus has been reached. In this study, an improved ADS was prepared by mesh technology, physicochemical experiments were conducted, cell adhesion and proliferation were assessed with the meshed ADS, and in vivo experiments were conducted to investigate the effects of meshed ADS or ADS combined with NPWT in repairing full-thickness skin defects. The results showed that the meshed ADS showed through-layer channels arranged in parallel longitudinal and transverse intersections. The cell experiments confirmed the good cytocompatibility. The in vivo experiments showed that there were no differences in the take rate or contraction of grafted skin among all experiment groups. The meshed ADS exhibited good histocompatibility, and there were no differences in tissue inflammation, dermal angiogenesis, or degradation among all groups. In addition, necrosis, dropsy, or empyema of the dermal scaffold were found in all experiment groups except for the meshed ADS + NPWT group, which showed better wound repair results, including fewer scaffold-related complications and satisfactory skin graft survival and wound contraction. In conclusion, this novel meshed ADS, which has a regular through-layer mesh structure and possesses stable physicochemical properties and good biocompatibility, combined with NPWT can ensure adequate subdermal drainage and reduce the risk of scaffold-related complications, thereby improving the quality and efficiency of wound repair, promoting a broader application of biomaterials, and helping physicians and readers implement more effective wound management.
Collapse
Affiliation(s)
- Pei Wei
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Lijiao Wu
- Department of Orthopedic Surgery, Fujian Provincial Hospital South Branch, Fuzhou 350001, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, China
| | - Hongteng Xie
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhaohong Chen
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Rongwei Tan
- GuangDong Engineering Technology Research Center of Implantable Medical Polymer, Shenzhen Lando Biomaterials Co., Ltd., Shenzhen 518107, China
| | - Zhaorong Xu
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| |
Collapse
|
13
|
Minne M, Terrie L, Wüst R, Hasevoets S, Vanden Kerchove K, Nimako K, Lambrichts I, Thorrez L, Declercq H. Generating human skeletal myoblast spheroids for vascular myogenic tissue engineering. Biofabrication 2024; 16:025035. [PMID: 38437715 DOI: 10.1088/1758-5090/ad2fd5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/04/2024] [Indexed: 03/06/2024]
Abstract
Engineered myogenic microtissues derived from human skeletal myoblasts offer unique opportunities for varying skeletal muscle tissue engineering applications, such asin vitrodrug-testing and disease modelling. However, more complex models require the incorporation of vascular structures, which remains to be challenging. In this study, myogenic spheroids were generated using a high-throughput, non-adhesive micropatterned surface. Since monoculture spheroids containing human skeletal myoblasts were unable to remain their integrity, co-culture spheroids combining human skeletal myoblasts and human adipose-derived stem cells were created. When using the optimal ratio, uniform and viable spheroids with enhanced myogenic properties were achieved. Applying a pre-vascularization strategy, through addition of endothelial cells, resulted in the formation of spheroids containing capillary-like networks, lumina and collagen in the extracellular matrix, whilst retaining myogenicity. Moreover, sprouting of endothelial cells from the spheroids when encapsulated in fibrin was allowed. The possibility of spheroids, from different maturation stages, to assemble into a more large construct was proven by doublet fusion experiments. The relevance of using three-dimensional microtissues with tissue-specific microarchitecture and increased complexity, together with the high-throughput generation approach, makes the generated spheroids a suitable tool forin vitrodrug-testing and human disease modeling.
Collapse
Affiliation(s)
- Mendy Minne
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Lisanne Terrie
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Rebecca Wüst
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Steffie Hasevoets
- Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, UHasselt, Diepenbeek, Belgium
| | - Kato Vanden Kerchove
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Kakra Nimako
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Ivo Lambrichts
- Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, UHasselt, Diepenbeek, Belgium
| | - Lieven Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Heidi Declercq
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| |
Collapse
|
14
|
Näf L, Miescher I, Pfuderer L, Schweizer TA, Brunner D, Dürig J, Gröninger O, Rieber J, Meier-Buergisser G, Spanaus K, Calcagni M, Bosshard PP, Achermann Y, Stark WJ, Buschmann J. Pro-angiogenic and antibacterial copper containing nanoparticles in PLGA/amorphous calcium phosphate bone nanocomposites. Heliyon 2024; 10:e27267. [PMID: 38486752 PMCID: PMC10937708 DOI: 10.1016/j.heliyon.2024.e27267] [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: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
Large bone defects after trauma demand for adequate bone substitutes. Bone void fillers should be antibacterial and pro-angiogenic. One viable option is the use of composite materials like the combination of PLGA and amorphous calcium phosphate (aCaP). Copper stimulates angiogenesis and has antibacterial qualities. Either copper oxide (CuO) nanoparticles (NPs) were therefore added to PLGA/aCaP/CuO in different concentrations (1, 5 and 10 w/w %) or copper-doped tricalcium phosphate NPs (TCP with 2% of copper) were electrospun into PLGA/CuTCP nanocomposites. Bi-layered nanocomposites of PLGA/aCaP with different copper NPs (CuO or TCP) and a second layer of pristine PLGA were fabricated. Two clinical bacterial isolates (Staphylococcus aureus and Staphylococcus epidermidis) were used to assess antibacterial properties of the copper-containing materials. For angiogenesis, the chorioallantoic membrane (CAM) assay of the chicken embryo was performed. The higher the CuO content, the higher were the antibacterial properties, with 10 % CuO reducing bacterial adhesion most effectively. Vessel and cell densities were highest in the 5 % CuO containing scaffolds, while tissue integration was more pronounced at lower CuO content. The PLGA/aCaP/CuO (1 % CuO) behaved similar like PLGA/CuTCP in all angiogenic and antibacterial readouts, based on the same copper fraction. We conclude that CuO NPs or CuTCP NPs are useful components to increase angiogenic properties of nanocomposites and at the same time exhibiting antibacterial characteristics.
Collapse
Affiliation(s)
- Lukas Näf
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Iris Miescher
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Lara Pfuderer
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Tiziano A. Schweizer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - David Brunner
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johannes Dürig
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Olivier Gröninger
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Julia Rieber
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Gabriella Meier-Buergisser
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Katharina Spanaus
- Clinical Chemistry, University Hospital Zurich, 8001, Zurich, Switzerland
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Philipp P. Bosshard
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Yvonne Achermann
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Wendelin J. Stark
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Johanna Buschmann
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
15
|
Yang J, Yan Y, Yin X, Liu X, Reshetov IV, Karalkin PA, Li Q, Huang RL. Bioengineering and vascularization strategies for islet organoids: advancing toward diabetes therapy. Metabolism 2024; 152:155786. [PMID: 38211697 DOI: 10.1016/j.metabol.2024.155786] [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: 10/23/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Diabetes presents a pressing healthcare crisis, necessitating innovative solutions. Organoid technologies have rapidly advanced, leading to the emergence of bioengineering islet organoids as an unlimited source of insulin-producing cells for treating insulin-dependent diabetes. This advancement surpasses the need for cadaveric islet transplantation. However, clinical translation of this approach faces two major limitations: immature endocrine function and the absence of a perfusable vasculature compared to primary human islets. In this review, we summarize the latest developments in bioengineering functional islet organoids in vitro and promoting vascularization of organoid grafts before and after transplantation. We highlight the crucial roles of the vasculature in ensuring long-term survival, maturation, and functionality of islet organoids. Additionally, we discuss key considerations that must be addressed before clinical translation of islet organoid-based therapy, including functional immaturity, undesired heterogeneity, and potential tumorigenic risks.
Collapse
Affiliation(s)
- Jing Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China
| | - Yuxin Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China
| | - Xiya Yin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China; Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, China
| | - Xiangqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China
| | - Igor V Reshetov
- Institute of Cluster Oncology, Sechenov First Moscow State Medical University, 127473 Moscow, Russia
| | - Pavel A Karalkin
- Institute of Cluster Oncology, Sechenov First Moscow State Medical University, 127473 Moscow, Russia
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China.
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China; Shanghai Institute for Plastic and Reconstructive Surgery, China.
| |
Collapse
|
16
|
Hauser PV, Zhao L, Chang HM, Yanagawa N, Hamon M. In Vivo Vascularization Chamber for the Implantation of Embryonic Kidneys. Tissue Eng Part C Methods 2024; 30:63-72. [PMID: 38062758 DOI: 10.1089/ten.tec.2023.0225] [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: 01/27/2024] Open
Abstract
A major obstacle to the implantation of ex vivo engineered tissues is the incorporation of functional vascular supply to support the growth of new tissue and to minimize ischemic injury. Existing prevascularization systems, such as arteriovenous (AV) loop-based systems, require microsurgery, limiting their use to larger animals. We aimed to develop an implantable device that can be prevascularized to enable vascularization of tissues in small rodents, and test its application on the vascularization of embryonic kidneys. Implanting the chamber between the abdominal aorta and the inferior vena cava, we detected endothelial cells and vascular networks after 48 h of implantation. Loading the chamber with collagen I (C), Matrigel (M), or Matrigel + vascular endothelial growth factor) (MV) had a strong influence on vascularization speed: Chambers loaded with C took 7 days to vascularize, 4 days for chambers with M, and 2 days for chambers with MV. Implantation of E12.5 mouse embryonic kidneys into prevascularized chambers (C, MV) was followed with significant growth and ureteric branching over 22 days. In contrast, the growth of kidneys in non-prevascularized chambers was stunted. We concluded that our prevascularized chamber is a valuable tool for vascularizing implanted tissues and tissue-engineered constructs. Further optimization will be necessary to control the directional growth of vascular endothelial cells within the chamber and the vascularization grade. Impact Statement Vascularization of engineered tissue, or organoids, constructs is a major hurdle in tissue engineering. Failure of vascularization is associated with prolonged ischemia time and potential tissue damage due to hypoxic effects. The method presented, demonstrates the use of a novel chamber that allows rapid vascularization of native and engineered tissues. We hope that this technology helps to stimulate research in the field of tissue vascularization and enables researchers to generate larger engineered vascularized tissues.
Collapse
Affiliation(s)
- Peter Viktor Hauser
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Lifu Zhao
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
| | - Hsiao-Min Chang
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Norimoto Yanagawa
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Morgan Hamon
- Division of Research, Renal Regeneration Laboratory, VAGLAHS at Sepulveda, North Hills, California, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| |
Collapse
|
17
|
Yang B, Yang G, Zhao F, Yao X, Xu L, Zhou L. Autologous Endothelial Progenitor Cells and Bioactive Factors Improve Bladder Regeneration. Tissue Eng Part C Methods 2024; 30:15-26. [PMID: 37756374 DOI: 10.1089/ten.tec.2023.0079] [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] [Indexed: 09/29/2023] Open
Abstract
Insufficient vascularization is still a challenge that impedes bladder tissue engineering and results in unsatisfied smooth muscle regeneration. Since bladder regeneration is a complex articulated process, the aim of this study is to investigate whether combining multiple pathways by exploiting a combination of biomaterials, cells, and bioactive factors, contributes to the improvements of smooth muscle regeneration and vascularization in tissue-engineered bladder. Autologous endothelial progenitor cells (EPCs) and bladder smooth muscle cells (BSMCs) are cultured and incorporated into our previously prepared porcine bladder acellular matrix (BAM) for bladder augmentation in rabbits. Simultaneously, exogenous vascular endothelial growth factor (VEGF) and platelet-derived growth factor BB (PDGF-BB) mixed with Matrigel were injected around the implanted cells-BAM complex. In the results, compared with control rabbits received bladder augmentation with porcine BAM seeded with BSMCs, the experimental animals showed significantly improved smooth muscle regeneration and vascularization, along with more excellent functional recovery of tissue-engineered bladder, due to the additional combination of autologous EPCs and bioactive factors, including VEGF and PDGF-BB. Furthermore, cell tracking suggested that the seeded EPCs could be directly involved in neovascularization. Therefore, it may be an effective method to combine multiple pathways for tissue-engineering urinary bladder.
Collapse
Affiliation(s)
- Bin Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guanjie Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Fellin CR, Steiner RC, Buchen JT, Anders JJ, Jariwala SH. Photobiomodulation and Vascularization in Conduit-Based Peripheral Nerve Repair: A Narrative Review. Photobiomodul Photomed Laser Surg 2024; 42:1-10. [PMID: 38109199 DOI: 10.1089/photob.2023.0103] [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: 12/20/2023] Open
Abstract
Background: Peripheral nerve injuries pose a significant clinical issue for patients, especially in the most severe cases wherein complete transection (neurotmesis) results in total loss of sensory/motor function. Nerve guidance conduits (NGCs) are a common treatment option that protects and guides regenerating axons during recovery. However, treatment outcomes remain limited and often fail to achieve full reinnervation, especially in critically sized defects (>3 cm) where a lack of vascularization leads to neural necrosis. Conclusions: A multitreatment approach is, therefore, necessary to improve the efficacy of NGCs. Stimulating angiogenesis within NGCs can help alleviate oxygen deficiency through rapid inosculation with the host vasculature, whereas photobiomodulation therapy (PBMT) has demonstrated beneficial therapeutic effects on regenerating nerve cells and neovascularization. In this review, we discuss the current trends of NGCs, vascularization, and PBMT as treatments for peripheral nerve neurotmesis and highlight the need for a combinatorial approach to improve functional and clinical outcomes.
Collapse
Affiliation(s)
- Christopher R Fellin
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- The Center for Rehabilitation Sciences Research, Department of Physical Medicine and Rehabilitation, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Richard C Steiner
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- The Center for Rehabilitation Sciences Research, Department of Physical Medicine and Rehabilitation, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Jack T Buchen
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- The Center for Rehabilitation Sciences Research, Department of Physical Medicine and Rehabilitation, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Juanita J Anders
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Shailly H Jariwala
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- The Center for Rehabilitation Sciences Research, Department of Physical Medicine and Rehabilitation, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Falkner F, Mayer SA, Heuer M, Brune J, Helt H, Bigdeli AK, Dimmler A, Heimel P, Thiele W, Sleeman JP, Bergmeister H, Schneider KH, Kneser U, Thomas B. Comparison of Decellularized Human Dermal Scaffolds versus Bovine Collagen/Elastin Matrices for Engineering of Soft-Tissue Flaps. Plast Reconstr Surg 2024; 153:130-141. [PMID: 37014963 DOI: 10.1097/prs.0000000000010511] [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: 04/06/2023]
Abstract
BACKGROUND Free flap-based soft-tissue reconstruction comes at the price of donor-site morbidity. The arteriovenous loop (AVL) technique can overcome this issue by allowing for the de novo generation of axially vascularized soft-tissue flaps from vein grafts embedded into different matrices. Application of the AVL technique has been limited by insufficient long-term volume retention and poor tissue stability. The authors investigated the suitability of a novel human dermal scaffold to improve volume retention and tissue stability. METHODS AVLs were created in 28 immunocompetent rats and embedded in either decellularized human dermal scaffolds (experimental group, n = 14) (Epiflex) or bovine collagen/elastin matrices (control group, n = 14) (MatriDerm) in subcutaneous polytetrafluoroethylene chambers. The weight and volume of engineered tissues, the extent of angiogenesis, and the proportion of proliferating cells were compared between groups on postoperative days (PODs) 21 and 28 by means of immunohistochemistry and micro-computed tomography. RESULTS On POD 28, both groups displayed homogeneous microvascular networks on histopathology and micro-computed tomography. Mean microvessel counts and surface areas and the percentage of proliferating cells did not differ between the groups. However, the experimental human scaffold group displayed significantly smaller volume loss and significantly less tissue degradation compared with bovine matrix controls (volume retention, 102% ± 5% versus 27% ± 7% on POD 21, and 79% ± 12% versus 12% ± 7% on POD 28, respectively; P < 0.0001). CONCLUSION Compared with bovine matrices, decellularized human scaffolds allow for superior volume retention and tissue stability of de novo engineered soft-tissue AVL flaps in rats. CLINICAL RELEVANCE STATEMENT AVLs allow for the de novo generation of vascularized soft-tissue flaps. However, insufficient long-term volume retention is still an issue. The authors' study shows that decellularized human matrices guarantee superior volume stability of de novo grown soft-tissue flaps in rats.
Collapse
Affiliation(s)
- Florian Falkner
- From the Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen
| | - Simon A Mayer
- From the Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen
| | - Miriam Heuer
- German Institute for Cell and Tissue Replacement
| | - Jan Brune
- German Institute for Cell and Tissue Replacement
| | - Hannah Helt
- From the Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen
| | - Amir K Bigdeli
- From the Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen
| | - Arno Dimmler
- Institute of Pathology, Vincentius Kliniken Karlsruhe
| | - Patrick Heimel
- Core Facility Hard Tissue and Biomaterial Research, Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology
| | - Wilko Thiele
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg
- Institute for Biological and Chemical Systems, Karlsruhe Institute of Technology, Campus North
| | - Jonathan P Sleeman
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg
- Institute for Biological and Chemical Systems, Karlsruhe Institute of Technology, Campus North
| | | | | | - Ulrich Kneser
- From the Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen
| | - Benjamin Thomas
- From the Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen
| |
Collapse
|
20
|
Zhao Z, Sun Y, Qiao Q, Weir MD, Schneider A, Masri R, Lynch CD, Zhang N, Zhang K, Bai Y, Xu H. Calvaria defect regeneration via human periodontal ligament stem cells and prevascularized scaffolds in athymic rats. J Dent 2023; 138:104690. [PMID: 37666466 DOI: 10.1016/j.jdent.2023.104690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Vascularization plays an important role in dental and craniofacial regenerations. Human periodontal ligament stem cells (hPDLSCs) are a promising cell source and, when co-cultured with human umbilical vein endothelial cells (hUVECs), could promote vascularization. The objectives of this study were to develop a novel prevascularized hPDLSC-hUVEC-calcium phosphate construct, and investigate the osteogenic and angiogenic efficacy of this construct with human platelet lysate (hPL) in cranial defects in rats for the first time. METHODS hPDLSCs and hUVECs were co-cultured on calcium phosphate cement (CPC) scaffolds with hPL. Cell proliferation, angiogenic gene expression, angiogenesis, alkaline phosphatase activity, and cell-synthesized minerals were determined. Bone and vascular regenerations were investigated in rat critical-sized cranial defects in vivo. RESULTS hPDLSC-hUVEC-CPC-hPL group had 2-fold greater angiogenic expressions and cell-synthesized mineral synthesis than hPDLSC-hUVEC-CPC group (p < 0.05). Microcapillary-like structures were formed on scaffolds in vitro. hPDLSC-hUVEC-CPC-hPL group had more vessels than hPDLSC-hUVEC-CPC group (p < 0.05). In cranial defects in rats, hPDLSC-hUVEC-CPC-hPL group regenerated new bone amount that was 2.1 folds and 4.0 folds, respectively, that of hPDLSC-hUVEC-CPC group and CPC control (p < 0.05). New blood vessel density of hPDLSC-hUVEC-CPC-hPL group was 2 folds and 7.9 folds, respectively, that of hPDLSC-hUVEC-CPC group and CPC control (p < 0.05). CONCLUSION The hPL pre-culture method is promising to enhance bone regeneration via prevascularized CPC. Novel hPDLSC-hUVEC-CPC-hPL prevascularized construct increased new bone formation and blood vessel density by 4-8 folds over CPC control. CLINICAL SIGNIFICANCE Novel hPDLSC-hUVEC-hPL-CPC prevascularized construct greatly increased bone and vascular regeneration in vivo and hence is promising for a wide range of craniofacial applications.
Collapse
Affiliation(s)
- Zeqing Zhao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yaxi Sun
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Radi Masri
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Christopher D Lynch
- Restorative Dentistry, University Dental School and Hospital, University College Cork, Wilton, Cork, Ireland
| | - Ning Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Hockin Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
21
|
Tong X, Xiao Z, Li P, Liu X, Wang M, Wen S, Wang N, Liao S, Zhou J. Angiogenesis and flap-related research: A bibliometric analysis. Int Wound J 2023; 20:3057-3072. [PMID: 37312275 PMCID: PMC10502283 DOI: 10.1111/iwj.14181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 06/15/2023] Open
Abstract
Adequate blood supply, a prerequisite for flap survival after grafting, makes angiogenesis of the flap the biggest problem to be solved. Researches have been conducted around vascularisation in correlation with flap grafting. However, bibliometric analyses systematically examining this research field are lacking. As such, we herein sought to conduct comprehensive comparative analyses of the contributions of different researchers, institutions, and countries to this research space in an effort to identify trends and hotspots in angiogenesis and vascularisation in the context of flap grafting. Publications pertaining to angiogenesis and vascularisation in the context of flap grafting were retrieved from the Web of Science Core Collection. References were then analysed and plotted using Microsoft Excel 2019, VOSviewer, and CiteSpace V. In total, 2234 papers that were cited 40 048 times (17.63 citations/paper) were included in this analysis. The greatest number of studies were from the United States, with these studies exhibiting both the highest number of citations (13 577) and the greatest overall H-index (60). For The institutions that published the greatest number of studies were WENZHOU MEDICAL UNIVERSITY (681), while UNIVERSITY OF ERLANGEN NUREMBERG has the highest number of citations (1458), and SHANGHAI JIAO TONG UNIVERSITY holds the greatest overall H-index (20). The greatest number of studies in this research space were published by Gao WY, while Horch RE was the most commonly cited researcher in the field. The VOS viewer software clustered relevant keywords into three clusters, with clusters 1, 2, 3, and 4 corresponding to studies in which the keywords 'anatomy', 'survival', 'transplantation', 'therapy' most frequently appeared. The most promising research hotspot-related terms in this field included 'autophagy', 'oxidative stress', 'ischemia/reperfusion injury', which exhibited a most recent average appearing year (AAY) of 2017 and after. Generally speaking, the results of this analysis indicate that the number of articles exploring angiogenesis and flap-related research has risen steadily, with the United States and China being the two countries publishing the greatest proportion of studies in this field. The overall focus of these studies has shifted away from 'infratest and tissue engineering' towards 'mechanisms'. In the future, particular attention should be paid to emerging research hotspots, which include 'ischemia/reperfusion injury' and treatments for promoting vascularization, such as 'platelet-rich plasma'. In light of these findings, funding agencies should continue increasing their investment in the exploration of the concrete mechanisms and interventional therapeutic relevance of angiogenesis during flap transplantation.
Collapse
Affiliation(s)
- Xiao‐Fei Tong
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Zhen‐Yang Xiao
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Pei‐Ting Li
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Xin Liu
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Ming‐Zhu Wang
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Shi‐Yi Wen
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Na Wang
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Shenghui Liao
- School of Computer Science and EngineeringCentral South UniversityChangshaChina
| | - Jian‐Da Zhou
- Department of Plastic SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
22
|
Hassan S, Wang T, Shi K, Huang Y, Urbina Lopez ME, Gan K, Chen M, Willemen N, Kalam H, Luna-Ceron E, Cecen B, Elbait GD, Li J, Garcia-Rivera LE, Gurian M, Banday MM, Yang K, Lee MC, Zhuang W, Johnbosco C, Jeon O, Alsberg E, Leijten J, Shin SR. Self-oxygenation of engineered living tissues orchestrates osteogenic commitment of mesenchymal stem cells. Biomaterials 2023; 300:122179. [PMID: 37315386 PMCID: PMC10330822 DOI: 10.1016/j.biomaterials.2023.122179] [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: 09/12/2022] [Revised: 04/12/2023] [Accepted: 05/25/2023] [Indexed: 06/16/2023]
Abstract
Oxygenating biomaterials can alleviate anoxic stress, stimulate vascularization, and improve engraftment of cellularized implants. However, the effects of oxygen-generating materials on tissue formation have remained largely unknown. Here, we investigate the impact of calcium peroxide (CPO)-based oxygen-generating microparticles (OMPs) on the osteogenic fate of human mesenchymal stem cells (hMSCs) under a severely oxygen deficient microenvironment. To this end, CPO is microencapsulated in polycaprolactone to generate OMPs with prolonged oxygen release. Gelatin methacryloyl (GelMA) hydrogels containing osteogenesis-inducing silicate nanoparticles (SNP hydrogels), OMPs (OMP hydrogels), or both SNP and OMP (SNP/OMP hydrogels) are engineered to comparatively study their effect on the osteogenic fate of hMSCs. OMP hydrogels associate with improved osteogenic differentiation under both normoxic and anoxic conditions. Bulk mRNAseq analyses suggest that OMP hydrogels under anoxia regulate osteogenic differentiation pathways more strongly than SNP/OMP or SNP hydrogels under either anoxia or normoxia. Subcutaneous implantations reveal a stronger host cell invasion in SNP hydrogels, resulting in increased vasculogenesis. Furthermore, time-dependent expression of different osteogenic factors reveals progressive differentiation of hMSCs in OMP, SNP, and SNP/OMP hydrogels. Our work demonstrates that endowing hydrogels with OMPs can induce, improve, and steer the formation of functional engineered living tissues, which holds potential for numerous biomedical applications, including tissue regeneration and organ replacement therapy.
Collapse
Affiliation(s)
- Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA; Department of Biology, College of Arts and Sciences, Khalifa University (Main Campus), Abu Dhabi, P.O. Box, 127788, United Arab Emirates; Advanced Materials Chemistry Center (AMCC), Khalifa University (SAN Campus), Abu Dhabi, P.O. Box, 127788, United Arab Emirates
| | - Ting Wang
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA; Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Kun Shi
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Maria Elizabeth Urbina Lopez
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Kaifeng Gan
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Mo Chen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Niels Willemen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA; Leijten Lab, Department of Developmental Bioengineering, Faculty of Science and Technology, TechMed Centre, University Twente, Enschede, 7522 NB, the Netherlands
| | - Haroon Kalam
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA
| | - Eder Luna-Ceron
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Berivan Cecen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Gihan Daw Elbait
- Department of Biology, College of Arts and Sciences, Khalifa University (Main Campus), Abu Dhabi, P.O. Box, 127788, United Arab Emirates
| | - Jinghang Li
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Luis Enrique Garcia-Rivera
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Melvin Gurian
- Leijten Lab, Department of Developmental Bioengineering, Faculty of Science and Technology, TechMed Centre, University Twente, Enschede, 7522 NB, the Netherlands
| | - Mudassir Meraj Banday
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Kisuk Yang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA; Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Weida Zhuang
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Castro Johnbosco
- Leijten Lab, Department of Developmental Bioengineering, Faculty of Science and Technology, TechMed Centre, University Twente, Enschede, 7522 NB, the Netherlands
| | - Oju Jeon
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60612, USA; Departments of Orthopaedic Surgery, Pharmacology and Regenerative Medicine, and Mechanical and Industrial Engineering, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Jeroen Leijten
- Leijten Lab, Department of Developmental Bioengineering, Faculty of Science and Technology, TechMed Centre, University Twente, Enschede, 7522 NB, the Netherlands.
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA.
| |
Collapse
|
23
|
Mohd N, Razali M, Fauzi MB, Abu Kasim NH. In Vitro and In Vivo Biological Assessments of 3D-Bioprinted Scaffolds for Dental Applications. Int J Mol Sci 2023; 24:12881. [PMID: 37629064 PMCID: PMC10454183 DOI: 10.3390/ijms241612881] [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: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Three-dimensional (3D) bioprinting is a unique combination of technological advances in 3D printing and tissue engineering. It has emerged as a promising approach to address the dilemma in current dental treatments faced by clinicians in order to repair or replace injured and diseased tissues. The exploration of 3D bioprinting technology provides high reproducibility and precise control of the bioink containing the desired cells and biomaterial over the architectural and dimensional features of the scaffolds in fabricating functional tissue constructs that are specific to the patient treatment need. In recent years, the dental applications of different 3D bioprinting techniques, types of novel bioinks, and the types of cells used have been extensively explored. Most of the findings noted significant challenges compared to the non-biological 3D printing approach in constructing the bioscaffolds that mimic native tissues. Hence, this review focuses solely on the implementation of 3D bioprinting techniques and strategies based on cell-laden bioinks. It discusses the in vitro applications of 3D-bioprinted scaffolds on cell viabilities, cell functionalities, differentiation ability, and expression of the markers as well as the in vivo evaluations of the implanted bioscaffolds on the animal models for bone, periodontal, dentin, and pulp tissue regeneration. Finally, it outlines some perspectives for future developments in dental applications.
Collapse
Affiliation(s)
- Nurulhuda Mohd
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Masfueh Razali
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Dean Office, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
24
|
Knabe C, Stiller M, Kampschulte M, Wilbig J, Peleska B, Günster J, Gildenhaar R, Berger G, Rack A, Linow U, Heiland M, Rendenbach C, Koerdt S, Steffen C, Houshmand A, Xiang-Tischhauser L, Adel-Khattab D. A tissue engineered 3D printed calcium alkali phosphate bioceramic bone graft enables vascularization and regeneration of critical-size discontinuity bony defects in vivo. Front Bioeng Biotechnol 2023; 11:1221314. [PMID: 37397960 PMCID: PMC10311449 DOI: 10.3389/fbioe.2023.1221314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction: Recently, efforts towards the development of patient-specific 3D printed scaffolds for bone tissue engineering from bioactive ceramics have continuously intensified. For reconstruction of segmental defects after subtotal mandibulectomy a suitable tissue engineered bioceramic bone graft needs to be endowed with homogenously distributed osteoblasts in order to mimic the advantageous features of vascularized autologous fibula grafts, which represent the standard of care, contain osteogenic cells and are transplanted with the respective blood vessel. Consequently, inducing vascularization early on is pivotal for bone tissue engineering. The current study explored an advanced bone tissue engineering approach combining an advanced 3D printing technique for bioactive resorbable ceramic scaffolds with a perfusion cell culture technique for pre-colonization with mesenchymal stem cells, and with an intrinsic angiogenesis technique for regenerating critical size, segmental discontinuity defects in vivo applying a rat model. To this end, the effect of differing Si-CAOP (silica containing calcium alkali orthophosphate) scaffold microarchitecture arising from 3D powder bed printing (RP) or the Schwarzwalder Somers (SSM) replica fabrication technique on vascularization and bone regeneration was analyzed in vivo. In 80 rats 6-mm segmental discontinuity defects were created in the left femur. Methods: Embryonic mesenchymal stem cells were cultured on RP and SSM scaffolds for 7d under perfusion to create Si-CAOP grafts with terminally differentiated osteoblasts and mineralizing bone matrix. These scaffolds were implanted into the segmental defects in combination with an arteriovenous bundle (AVB). Native scaffolds without cells or AVB served as controls. After 3 and 6 months, femurs were processed for angio-µCT or hard tissue histology, histomorphometric and immunohistochemical analysis of angiogenic and osteogenic marker expression. Results: At 3 and 6 months, defects reconstructed with RP scaffolds, cells and AVB displayed a statistically significant higher bone area fraction, blood vessel volume%, blood vessel surface/volume, blood vessel thickness, density and linear density than defects treated with the other scaffold configurations. Discussion: Taken together, this study demonstrated that the AVB technique is well suited for inducing adequate vascularization of the tissue engineered scaffold graft in segmental defects after 3 and 6 months, and that our tissue engineering approach employing 3D powder bed printed scaffolds facilitated segmental defect repair.
Collapse
Affiliation(s)
- Christine Knabe
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
| | - Michael Stiller
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
- Department of Prosthodontics, Philipps University Marburg, Marburg, Germany
| | - Marian Kampschulte
- Department of Radiology, Justus Liebig University Giessen, Giessen, Germany
| | - Janka Wilbig
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Barbara Peleska
- Department of Prosthodontics, Philipps University Marburg, Marburg, Germany
| | - Jens Günster
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Renate Gildenhaar
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Georg Berger
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Alexander Rack
- Structure of Materials Group, ESRF (European Synchroton Radiation Facility), Grenoble, France
| | - Ulf Linow
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Rendenbach
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Steffen Koerdt
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudius Steffen
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alireza Houshmand
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
| | - Li Xiang-Tischhauser
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
| | - Doaa Adel-Khattab
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
- Department of Periodontology, Ain Shams University, Cairo, Egypt
| |
Collapse
|
25
|
Tang X, Ren J, Wei X, Wang T, Li H, Sun Y, Liu Y, Chi M, Zhu S, Lu L, Zhang J, Yang B. Exploiting synergistic effect of CO/NO gases for soft tissue transplantation using a hydrogel patch. Nat Commun 2023; 14:2417. [PMID: 37105981 PMCID: PMC10140290 DOI: 10.1038/s41467-023-37959-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Autologous skin flap transplantation is a common method for repairing complex soft tissue defects caused by cancer, trauma, and congenital malformations. Limited blood supply range and post-transplantation ischemia-reperfusion injury can lead to distal necrosis of the flap and long-term functional loss, which severely restricts the decision-making regarding the optimal surgical plan. To address this issue, we develop a hydrogel patch that releases carbon monoxide and nitric oxide gases on demand, to afford a timely blood supply for skin flap transplantation during surgery. Using an ischemia-reperfusion dorsal skin flap model in rats, we show that the hydrogel patch maintains the immediate opening of blood flow channels in transplanted tissue and effective blood perfusion throughout the perioperative period, activating perfusion of the hemodynamic donor site. We demonstrate that the hydrogel patch promotes distal vascularization and long-term functional reconstruction of transplanted tissues by inhibiting inflammatory damage and accelerating blood vessel formation.
Collapse
Affiliation(s)
- Xiaoduo Tang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| | - Jingyan Ren
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| | - Xin Wei
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Tao Wang
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Haiqiu Li
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Yihan Sun
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| | - Yang Liu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Mingli Chi
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China.
| | - Laijin Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China.
| | - Junhu Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China.
| | - Bai Yang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| |
Collapse
|
26
|
Chen J, Zhang D, Wu LP, Zhao M. Current Strategies for Engineered Vascular Grafts and Vascularized Tissue Engineering. Polymers (Basel) 2023; 15:polym15092015. [PMID: 37177162 PMCID: PMC10181238 DOI: 10.3390/polym15092015] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Blood vessels not only transport oxygen and nutrients to each organ, but also play an important role in the regulation of tissue regeneration. Impaired or occluded vessels can result in ischemia, tissue necrosis, or even life-threatening events. Bioengineered vascular grafts have become a promising alternative treatment for damaged or occlusive vessels. Large-scale tubular grafts, which can match arteries, arterioles, and venules, as well as meso- and microscale vasculature to alleviate ischemia or prevascularized engineered tissues, have been developed. In this review, materials and techniques for engineering tubular scaffolds and vasculature at all levels are discussed. Examples of vascularized tissue engineering in bone, peripheral nerves, and the heart are also provided. Finally, the current challenges are discussed and the perspectives on future developments in biofunctional engineered vessels are delineated.
Collapse
Affiliation(s)
- Jun Chen
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Center for Chemical Biology and Drug Discovery, Laboratory of Computational Biomedicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Di Zhang
- Center for Chemical Biology and Drug Discovery, Laboratory of Computational Biomedicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Lin-Ping Wu
- Center for Chemical Biology and Drug Discovery, Laboratory of Computational Biomedicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ming Zhao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
27
|
Derman ID, Singh YP, Saini S, Nagamine M, Banerjee D, Ozbolat IT. Bioengineering and Clinical Translation of Human Lung and its Components. Adv Biol (Weinh) 2023; 7:e2200267. [PMID: 36658734 PMCID: PMC10121779 DOI: 10.1002/adbi.202200267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/18/2022] [Indexed: 01/21/2023]
Abstract
Clinical lung transplantation has rapidly established itself as the gold standard of treatment for end-stage lung diseases in a restricted group of patients since the first successful lung transplant occurred. Although significant progress has been made in lung transplantation, there are still numerous obstacles on the path to clinical success. The development of bioartificial lung grafts using patient-derived cells may serve as an alternative treatment modality; however, challenges include developing appropriate scaffold materials, advanced culture strategies for lung-specific multiple cell populations, and fully matured constructs to ensure increased transplant lifetime following implantation. This review highlights the development of tissue-engineered tracheal and lung equivalents over the past two decades, key problems in lung transplantation in a clinical environment, the advancements made in scaffolds, bioprinting technologies, bioreactors, organoids, and organ-on-a-chip technologies. The review aims to fill the lacuna in existing literature toward a holistic bioartificial lung tissue, including trachea, capillaries, airways, bifurcating bronchioles, lung disease models, and their clinical translation. Herein, the efforts are on bridging the application of lung tissue engineering methods in a clinical environment as it is thought that tissue engineering holds enormous promise for overcoming the challenges associated with the clinical translation of bioengineered human lung and its components.
Collapse
Affiliation(s)
- I. Deniz Derman
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
| | - Yogendra Pratap Singh
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
| | - Shweta Saini
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, India
| | - Momoka Nagamine
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
- Department of Chemistry, Penn State University; University Park, PA,16802, USA
| | - Dishary Banerjee
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
- Biomedical Engineering Department, Penn State University; University Park, PA, 16802, USA
- Materials Research Institute, Penn State University; University Park, PA, 16802, USA
- Cancer Institute, Penn State University; University Park, PA, 16802, USA
- Neurosurgery Department, Penn State University; University Park, PA, 16802, USA
- Department of Medical Oncology, Cukurova University, Adana, Turkey
| |
Collapse
|
28
|
Zhang L, Wang M, Qiu H, Wei Y, Zhou L, Nian N, Shi Z, Hu D, Ma B. Epicatechin gallate promotes vascularization in co-culture of human osteoblasts and outgrowth endothelial cells. Exp Biol Med (Maywood) 2023; 248:732-745. [PMID: 37354086 PMCID: PMC10408553 DOI: 10.1177/15353702231171894] [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: 11/24/2022] [Accepted: 02/27/2023] [Indexed: 06/26/2023] Open
Abstract
Prevascularization is crucial for the survival of tissue-engineered bone and further bone repair/regeneration. Since epicatechin gallate (ECG), the most abundant flavanol in green tea, shows potential beneficial effects on endothelial cells and bone cells, we decided to investigate whether it promotes vascularization/angiogenesis and osteogenesis using a co-culture system containing human primary osteoblasts (POBs) and outgrowth endothelial cells (OECs). We found that treatment with ECG (1) significantly enhanced microvessel formation in co-culture of POB and OECs, (2) improved cell viability/proliferation and the angiogenic/osteogenic capacities of OEC/POBs, (3) significantly increased the levels of E-selectin, IL-6, TNF-α, IFN-γ, VEGF, and PDGF-BB in co-cultures of POB and OEC, and (4) upregulated HIF-1α, HIF-2α, NF-κB, iNOS, GLUT1, VEGF, and Ang1/2 but downregulated PHD1 in monocultures of OEC or POB. Our findings demonstrate that ECG promotes angiogenesis and osteogenesis (probably via HIF signaling) in co-cultures of OECs and POBs. ECG thus has potential applications in the promotion of angiogenesis/vascularization in many tissue constructs including those of bone.
Collapse
Affiliation(s)
- Liyan Zhang
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Miaoran Wang
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Huiqing Qiu
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
- Department of Geriatrics, The First Hospital of Hebei Medical University, Shijiazhuang 050030, China
| | - Yusen Wei
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Lu Zhou
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Nannan Nian
- Graduate School, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhongli Shi
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, The First Hospital of Hebei Medical University, Shijiazhuang 050030, China
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang 050030, China
| | - Dailun Hu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang 050017, China
| | - Bin Ma
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
29
|
Dienemann S, Schmidt V, Fleischhammer T, Mueller JH, Lavrentieva A. Comparative analysis of hypoxic response of human microvascular and umbilical vein endothelial cells in 2D and 3D cell culture systems. J Cell Physiol 2023; 238:1111-1120. [PMID: 36947660 DOI: 10.1002/jcp.31002] [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: 12/22/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/24/2023]
Abstract
In vitro cultivation conditions play a crucial role in cell physiology and the cellular response to external stimuli. Oxygen concentrations represent an essential microenvironmental factor influencing cell physiology and behaviour both in vivo and in vitro. Therefore, new approaches are urgently needed to monitor and control oxygen concentrations in 2D and 3D cultures, as well as cell reactions to these concentrations. In this work, we modified two types of human endothelial cells-human microvascular (huMECs) and umbilical vein endothelial cells (huVECs) with genetically encoded hypoxia biosensors and monitored cell reactions in 2D to different oxygen concentrations. Moreover, we fabricated 3D cell spheroids of different cell numbers and sizes to reveal the onset of hypoxia in huVECs and huMECs. We could demonstrate a quantitative sensor response of two cell types to reduced oxygen supply in 2D and reveal different thresholds for hypoxic response. In 3D cell spheroids we could estimate critical construct sizes for the appearance of a hypoxic core. This work for the first time directly demonstrates different hypoxic signatures for huVECs and huMECs in 2D and 3D cell culture systems.
Collapse
Affiliation(s)
- Sandra Dienemann
- Institute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Vanessa Schmidt
- Institute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Tabea Fleischhammer
- Institute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Julia H Mueller
- Institute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| |
Collapse
|
30
|
Li A, Sasaki JI, Abe GL, Katata C, Sakai H, Imazato S. Vascularization of a Bone Organoid Using Dental Pulp Stem Cells. Stem Cells Int 2023; 2023:5367887. [PMID: 37200632 PMCID: PMC10188257 DOI: 10.1155/2023/5367887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023] Open
Abstract
Bone organoids offer a novel path for the reconstruction and repair of bone defects. We previously fabricated scaffold-free bone organoids using cell constructs comprising only bone marrow-derived mesenchymal stem cells (BMSCs). However, the cells in the millimetre-scale constructs were likely to undergo necrosis because of difficult oxygen diffusion and nutrient delivery. Dental pulp stem cells (DPSCs) are capable of differentiating into vascular endothelial lineages and have great vasculogenic potential under endothelial induction. Therefore, we hypothesized that DPSCs can serve as a vascular source to improve the survival of the BMSCs within the bone organoid. In this study, the DPSCs had greater sprouting ability, and the proangiogenic marker expressions were significantly greater than those of BMSCs. DPSCs were incorporated into the BMSC constructs at various ratios (5%-20%), and their internal structures and vasculogenic and osteogenic characteristics were investigated after endothelial differentiation. As a result, the DPSCs are differentiated into the CD31-positive endothelial lineage in the cell constructs. The incorporation of DPSCs significantly suppressed cell necrosis and improved the viability of the cell constructs. In addition, lumen-like structures were visualized by fluorescently labelled nanoparticles in the DPSC-incorporated cell constructs. The vascularized BMSC constructs were successfully fabricated using the vasculogenic ability of the DPSCs. Next, osteogenic induction was initiated in the vascularized BMSC/DPSC constructs. Compared with only BMSCs, constructs with DPSCs had increased mineralized deposition and a hollow structure. Overall, this study demonstrated that vascularized scaffold-free bone organoids were successfully fabricated by incorporating DPSCs into BMSC constructs, and the biomimetic biomaterial is promising for bone regenerative medicine and drug development.
Collapse
Affiliation(s)
- Aonan Li
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Jun-Ichi Sasaki
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Gabriela L. Abe
- Department of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Chihiro Katata
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hirohiko Sakai
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Satoshi Imazato
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
31
|
Oudda S, Ali AM, Chien AL, Park S. Leveraging Tissue Engineering for Skin Cancer Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:97-113. [PMID: 36484897 DOI: 10.1007/5584_2022_755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bioengineered in vitro three-dimensional (3D) skin model has emerged as a promising tool for recapitulating different types of skin cancer and performing pre-clinical tests. However, a full-thickness 3D model including the epidermis, dermis, and hypodermis layers is scarce despite its significance in human physiology and diverse biological processes. In this book chapter, an attempt has been made to summarize various skin cancer models, including utilized skin layers, materials, cell lines, specific treatments, and fabrication techniques for three types of skin cancer: melanoma, basal cell carcinoma (BCC), and squamous cell carcinoma (SCC). Subsequently, current limitations and future directions of skin cancer models are discussed. The knowledge of the current status of skin cancer models can provide various potential applications in cancer research and thus a more effective way for cancer treatment.
Collapse
Affiliation(s)
- Sumayah Oudda
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Abdulla M Ali
- The Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD, USA
| | - Anna L Chien
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seungman Park
- Department of Mechanical Engineering, University of Nevada, Las Vegas, Las Vegas, NV, USA.
| |
Collapse
|
32
|
Zhang Y, Luo W, Zheng L, Hu J, Nie L, Zeng H, Tan X, Jiang Y, Li Y, Zhao T, Yang Z, He TC, Zhang H. Efficient bone regeneration of BMP9-stimulated human periodontal ligament stem cells (hPDLSCs) in decellularized bone matrix (DBM) constructs to model maxillofacial intrabony defect repair. Stem Cell Res Ther 2022; 13:535. [PMID: 36575551 PMCID: PMC9795631 DOI: 10.1186/s13287-022-03221-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND BMP9-stimulated DPSCs, SCAPs and PDLSCs are effective candidates for repairing maxillofacial bone defects in tissue engineering, while the most suitable seed cell source among these three hDMSCs and the optimal combination of most suitable type of hDMSCs and BMP9 have rarely been explored. Moreover, the orthotopic maxillofacial bone defect model should be valuable but laborious and time-consuming to evaluate various candidates for bone regeneration. Thus, inspired from the maxillofacial bone defects and the traditional in vivo ectopic systems, we developed an intrabony defect repair model to recapitulate the healing events of orthotopic maxillofacial bone defect repair and further explore the optimized combinations of most suitable hDMSCs and BMP9 for bone defect repair based on this modified ectopic system. METHODS Intrabony defect repair model was developed by using decellularized bone matrix (DBM) constructs prepared from the cancellous part of porcine lumbar vertebral body. We implanted DBM constructs subcutaneously on the flank of each male NU/NU athymic nude mouse, followed by directly injecting the cell suspension of different combinations of hDMSCs and BMP9 into the central hollow area of the constructs 7 days later. Then, the quality of the bony mass, including bone volume fraction (BV/TV), radiographic density (in Hounsfield units (HU)) and the height of newly formed bone, was measured by micro-CT. Furthermore, the H&E staining and immunohistochemical staining were performed to exam new bone and new blood vessel formation in DBM constructs. RESULTS BMP9-stimulated periodontal ligament stem cells (PDLSCs) exhibited the most effective bone regeneration among the three types of hDMSCs in DBM constructs. Furthermore, an optimal dose of PDLSCs with a specific extent of BMP9 stimulation was confirmed for efficacious new bone and new blood vessel formation in DBM constructs. CONCLUSIONS The reported intrabony defect repair model can be used to identify optimized combinations of suitable seed cells and biological factors for bone defect repair and subsequent development of efficacious bone tissue engineering therapies.
Collapse
Affiliation(s)
- Yuxin Zhang
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Wenping Luo
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Liwen Zheng
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jing Hu
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Li Nie
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Huan Zeng
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Xi Tan
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Yucan Jiang
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Yeming Li
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Tianyu Zhao
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Zhuohui Yang
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Tong-Chuan He
- grid.412578.d0000 0000 8736 9513Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Hongmei Zhang
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, 426 Songshibei Road, Chongqing, 401147 China ,grid.203458.80000 0000 8653 0555Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Anthon SG, Valente KP. Vascularization Strategies in 3D Cell Culture Models: From Scaffold-Free Models to 3D Bioprinting. Int J Mol Sci 2022; 23:14582. [PMID: 36498908 PMCID: PMC9737506 DOI: 10.3390/ijms232314582] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
The discrepancies between the findings in preclinical studies, and in vivo testing and clinical trials have resulted in the gradual decline in drug approval rates over the past decades. Conventional in vitro drug screening platforms employ two-dimensional (2D) cell culture models, which demonstrate inaccurate drug responses by failing to capture the three-dimensional (3D) tissue microenvironment in vivo. Recent advancements in the field of tissue engineering have made possible the creation of 3D cell culture systems that can accurately recapitulate the cell-cell and cell-extracellular matrix interactions, as well as replicate the intricate microarchitectures observed in native tissues. However, the lack of a perfusion system in 3D cell cultures hinders the establishment of the models as potential drug screening platforms. Over the years, multiple techniques have successfully demonstrated vascularization in 3D cell cultures, simulating in vivo-like drug interactions, proposing the use of 3D systems as drug screening platforms to eliminate the deviations between preclinical and in vivo testing. In this review, the basic principles of 3D cell culture systems are briefly introduced, and current research demonstrating the development of vascularization in 3D cell cultures is discussed, with a particular focus on the potential of these models as the future of drug screening platforms.
Collapse
Affiliation(s)
- Shamapto Guha Anthon
- Department of Biomedical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | |
Collapse
|
34
|
Co-transplantation of pancreatic islets and microvascular fragments effectively restores normoglycemia in diabetic mice. NPJ Regen Med 2022; 7:67. [PMCID: PMC9636251 DOI: 10.1038/s41536-022-00262-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
AbstractInsufficient revascularization of pancreatic islets is one of the major obstacles impairing the success of islet transplantation. To overcome this problem, we introduce in the present study a straightforward strategy to accelerate the engraftment of isolated islets. For this purpose, we co-transplanted 250 islets and 20,000 adipose tissue-derived microvascular fragments (MVF) from donor mice under the kidney capsule as well as 500 or 1000 islets with 40,000 MVF into the subcutaneous space of diabetic mice. We found that the co-transplantation of islets and MVF markedly accelerates the restoration of normoglycemia in diabetic recipients compared with the transplantation of islets alone. In fact, the transplantation of 250 islets with 20,000 MVF under the kidney capsule reversed diabetes in 88% of mice and the subcutaneous transplantation of 500 or 1000 islets with 40,000 MVF restored normoglycemia in 100% of mice. Moreover, diabetic mice receiving islets and MVF exhibited plasma insulin levels similar to nondiabetic control animals. Additional immunohistochemical analyses of the grafts revealed a significantly higher number of islet cells and microvessels in the co-transplantation groups. These findings demonstrate that the co-transplantation of islets and MVF is a promising strategy to improve the success rates of islet transplantation, which could be easily implemented into future clinical practice.
Collapse
|
35
|
Grilli F, Pitton M, Altomare L, Farè S. Decellularized fennel and dill leaves as possible 3D channel network in GelMA for the development of an in vitro adipose tissue model. Front Bioeng Biotechnol 2022; 10:984805. [DOI: 10.3389/fbioe.2022.984805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
The development of 3D scaffold-based models would represent a great step forward in cancer research, offering the possibility of predicting the potential in vivo response to targeted anticancer or anti-angiogenic therapies. As regards, 3D in vitro models require proper materials, which faithfully recapitulated extracellular matrix (ECM) properties, adequate cell lines, and an efficient vascular network. The aim of this work is to investigate the possible realization of an in vitro 3D scaffold-based model of adipose tissue, by incorporating decellularized 3D plant structures within the scaffold. In particular, in order to obtain an adipose matrix capable of mimicking the composition of the adipose tissue, methacrylated gelatin (GelMA), UV photo-crosslinkable, was selected. Decellularized fennel, wild fennel and, dill leaves have been incorporated into the GelMA hydrogel before crosslinking, to mimic a 3D channel network. All leaves showed a loss of pigmentation after the decellularization with channel dimensions ranging from 100 to 500 µm up to 3 μm, comparable with those of human microcirculation (5–10 µm). The photo-crosslinking process was not affected by the embedded plant structures in GelMA hydrogels. In fact, the weight variation test, performed on hydrogels with or without decellularized leaves showed a weight loss in the first 96 h, followed by a stability plateau up to 5 weeks. No cytotoxic effects were detected comparing the three prepared GelMA/D-leaf structures; moreover, the ability of the samples to stimulate differentiation of 3T3-L1 preadipocytes in mature adipocytes was investigated, and cells were able to grow and proliferate in the structure, colonizing the entire microenvironment and starting to differentiate. The developed GelMA hydrogels mimicked adipose tissue together with the incorporated plant structures seem to be an adequate solution to ensure an efficient vascular system for a 3D in vitro model. The obtained results showed the potentiality of the innovative proposed approach to mimic the tumoral microenvironment in 3D scaffold-based models.
Collapse
|
36
|
Tsiklin IL, Shabunin AV, Kolsanov AV, Volova LT. In Vivo Bone Tissue Engineering Strategies: Advances and Prospects. Polymers (Basel) 2022; 14:polym14153222. [PMID: 35956735 PMCID: PMC9370883 DOI: 10.3390/polym14153222] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/25/2022] [Accepted: 08/04/2022] [Indexed: 12/12/2022] Open
Abstract
Reconstruction of critical-sized bone defects remains a tremendous challenge for surgeons worldwide. Despite the variety of surgical techniques, current clinical strategies for bone defect repair demonstrate significant limitations and drawbacks, including donor-site morbidity, poor anatomical match, insufficient bone volume, bone graft resorption, and rejection. Bone tissue engineering (BTE) has emerged as a novel approach to guided bone tissue regeneration. BTE focuses on in vitro manipulations with seed cells, growth factors and bioactive scaffolds using bioreactors. The successful clinical translation of BTE requires overcoming a number of significant challenges. Currently, insufficient vascularization is the critical limitation for viability of the bone tissue-engineered construct. Furthermore, efficacy and safety of the scaffolds cell-seeding and exogenous growth factors administration are still controversial. The in vivo bioreactor principle (IVB) is an exceptionally promising concept for the in vivo bone tissue regeneration in a predictable patient-specific manner. This concept is based on the self-regenerative capacity of the human body, and combines flap prefabrication and axial vascularization strategies. Multiple experimental studies on in vivo BTE strategies presented in this review demonstrate the efficacy of this approach. Routine clinical application of the in vivo bioreactor principle is the future direction of BTE; however, it requires further investigation for overcoming some significant limitations.
Collapse
Affiliation(s)
- Ilya L. Tsiklin
- Biotechnology Center “Biotech”, Samara State Medical University, 443079 Samara, Russia
- City Clinical Hospital Botkin, Moscow Healthcare Department, 125284 Moscow, Russia
- Correspondence: ; Tel.: +7-903-621-81-88
| | - Aleksey V. Shabunin
- City Clinical Hospital Botkin, Moscow Healthcare Department, 125284 Moscow, Russia
| | - Alexandr V. Kolsanov
- Biotechnology Center “Biotech”, Samara State Medical University, 443079 Samara, Russia
| | - Larisa T. Volova
- Biotechnology Center “Biotech”, Samara State Medical University, 443079 Samara, Russia
| |
Collapse
|
37
|
Petta D, D'Amora U, D'Arrigo D, Tomasini M, Candrian C, Ambrosio L, Moretti M. Musculoskeletal tissues-on-a-chip: role of natural polymers in reproducing tissue-specific microenvironments. Biofabrication 2022; 14. [PMID: 35931043 DOI: 10.1088/1758-5090/ac8767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/05/2022] [Indexed: 11/12/2022]
Abstract
Over the past years, 3D in vitro models have been widely employed in the regenerative medicine field. Among them, organ-on-a-chip technology has the potential to elucidate cellular mechanism exploiting multichannel microfluidic devices to establish 3D co-culture systems that offer control over the cellular, physico-chemical and biochemical microenvironments. To deliver the most relevant cues to cells, it is of paramount importance to select the most appropriate matrix for mimicking the extracellular matrix of the native tissue. Natural polymers-based hydrogels are the elected candidates for reproducing tissue-specific microenvironments in musculoskeletal tissue-on-a-chip models owning to their interesting and peculiar physico-chemical, mechanical and biological properties. Despite these advantages, there is still a gap between the biomaterials complexity in conventional tissue engineering and the application of these biomaterials in 3D in vitro microfluidic models. In this review, the aim is to suggest the adoption of more suitable biomaterials, alternative crosslinking strategies and tissue engineered-inspired approaches in organ-on-a-chip to better mimic the complexity of physiological musculoskeletal tissues. Accordingly, after giving an overview of the musculoskeletal tissue compositions, the properties of the main natural polymers employed in microfluidic systems are investigated, together with the main musculoskeletal tissues-on-a-chip devices.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologis Lab, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Ugo D'Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, V.le J.F. Kennedy 54 Mostra d'Oltremare Pad 20, Naples, 80125, ITALY
| | - Daniele D'Arrigo
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Marta Tomasini
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco chies 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Christian Candrian
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, via Tesserete 46, Lugano, 6900, SWITZERLAND
| | - Luigi Ambrosio
- Institute of Polymers Composites and Biomaterials National Research Council, Viale Kennedy, Pozzuoli, Campania, 80078, ITALY
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| |
Collapse
|
38
|
Liu G, Zhang B, Wan T, Zhou C, Fan Y, Tian W, Jing W. A 3D-printed biphasic calcium phosphate scaffold loaded with platelet lysate/gelatin methacrylate to promote vascularization. J Mater Chem B 2022; 10:3138-3151. [PMID: 35352743 DOI: 10.1039/d2tb00006g] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
3D-printed biphasic calcium phosphate (BCP) scaffolds show great clinical application potential in bone tissue engineering; however, vascularization of the scaffold is a crucial step for bone regeneration and is still difficult to be controlled. To enhance scaffold vascularization, a novel bioactive scaffold loaded with platelet lysate/gelatin methacrylate (PL/GelMA) in a BCP scaffold was proposed for promoting vascularization. The PL/GelMA/BCP scaffold was successfully prepared via digital light processing (DLP) printing and filled with PL/GelMA to promote the vascularization effect. In vitro evaluation indicated that human umbilical vein endothelial cells (HUVECs) adhered well on the PL/GelMA/BCP scaffold, and cell proliferation was significantly promoted by coculture with the scaffold. Moreover, a variety of growth factors (GFs) in the PL were detected which were slowly released from the scaffold to modulate the cell behaviour and promote the formation of blood vessel-like structures. Co-culturing with the PL/GelMA/BCP scaffold upregulated the expression of angiogenesis-related genes in cells. In vitro results showed that a higher capillary formation was also observed in PL/GelMA/BCP scaffolds implanted subcutaneously on the back of the rats. These results indicated that the vascularization ability of BCP was enhanced by filling it with PL/GelMA. The PL/GelMA/BCP scaffold has the potential to promote vascularization in tissue engineering.
Collapse
Affiliation(s)
- Gang Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China. .,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14, 3Rd Section of Ren Min Nan Rd., Chengdu, Sichuan 610041, China
| | - Boqing Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Ting Wan
- Affiliated Hospital of Sichuan Nursing Vocational College (The Third People's Hospital of Sichuan Province), Chengdu 610071, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China. .,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14, 3Rd Section of Ren Min Nan Rd., Chengdu, Sichuan 610041, China
| | - Wei Jing
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China. .,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14, 3Rd Section of Ren Min Nan Rd., Chengdu, Sichuan 610041, China
| |
Collapse
|
39
|
Ze Y, Li Y, Huang L, Shi Y, Li P, Gong P, Lin J, Yao Y. Biodegradable Inks in Indirect Three-Dimensional Bioprinting for Tissue Vascularization. Front Bioeng Biotechnol 2022; 10:856398. [PMID: 35402417 PMCID: PMC8990266 DOI: 10.3389/fbioe.2022.856398] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023] Open
Abstract
Mature vasculature is important for the survival of bioengineered tissue constructs, both in vivo and in vitro; however, the fabrication of fully vascularized tissue constructs remains a great challenge in tissue engineering. Indirect three-dimensional (3D) bioprinting refers to a 3D printing technique that can rapidly fabricate scaffolds with controllable internal pores, cavities, and channels through the use of sacrificial molds. It has attracted much attention in recent years owing to its ability to create complex vascular network-like channels through thick tissue constructs while maintaining endothelial cell activity. Biodegradable materials play a crucial role in tissue engineering. Scaffolds made of biodegradable materials act as temporary templates, interact with cells, integrate with native tissues, and affect the results of tissue remodeling. Biodegradable ink selection, especially the choice of scaffold and sacrificial materials in indirect 3D bioprinting, has been the focus of several recent studies. The major objective of this review is to summarize the basic characteristics of biodegradable materials commonly used in indirect 3D bioprinting for vascularization, and to address recent advances in applying this technique to the vascularization of different tissues. Furthermore, the review describes how indirect 3D bioprinting creates blood vessels and vascularized tissue constructs by introducing the methodology and biodegradable ink selection. With the continuous improvement of biodegradable materials in the future, indirect 3D bioprinting will make further contributions to the development of this field.
Collapse
Affiliation(s)
- Yiting Ze
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyang Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixin Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Zheng W, Xie R, Liang X, Liang Q. Fabrication of Biomaterials and Biostructures Based On Microfluidic Manipulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105867. [PMID: 35072338 DOI: 10.1002/smll.202105867] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Biofabrication technologies are of importance for the construction of organ models and functional tissue replacements. Microfluidic manipulation, a promising biofabrication technique with micro-scale resolution, can not only help to realize the fabrication of specific microsized structures but also build biomimetic microenvironments for biofabricated tissues. Therefore, microfluidic manipulation has attracted attention from researchers in the manipulation of particles and cells, biochemical analysis, tissue engineering, disease diagnostics, and drug discovery. Herein, biofabrication based on microfluidic manipulation technology is reviewed. The application of microfluidic manipulation technology in the manufacturing of biomaterials and biostructures with different dimensions and the control of the microenvironment is summarized. Finally, current challenges are discussed and a prospect of microfluidic manipulation technology is given. The authors hope this review can provide an overview of microfluidic manipulation technologies used in biofabrication and thus steer the current efforts in this field.
Collapse
Affiliation(s)
- Wenchen Zheng
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Ruoxiao Xie
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xiaoping Liang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangdong, 510006, China
| | - Qionglin Liang
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
41
|
Wu W, Luo H, Wu D, Shi M, Yu J, Liao H. Biological activity of a vascular endothelial cell-hydroxyapatite orbital implant complex: An experimental study. Exp Ther Med 2022; 23:227. [PMID: 35222704 PMCID: PMC8815058 DOI: 10.3892/etm.2022.11152] [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: 08/13/2021] [Accepted: 12/17/2021] [Indexed: 11/06/2022] Open
Abstract
The reduction in postoperative complications is a considerable concern after orbital reparation and reconstruction. Selecting the appropriate scaffold materials to improve the survival rates of the seeded cells is a challenge in tissue engineering. The aim of the present study was to evaluate the biological activity of a vascular endothelial cell-hydroxyapatite orbital complex, which was constructed with tissue engineering and used as an implant after enucleation of the eyeNew Zealand white rabbits were randomly divided into two groups that underwent hydroxyapatite orbital implant surgery in the right eye. The primary orbital microvascular endothelial cells were collected from the microvascular tissue and subsequently cultured. Then, hydroxyapatite ocular implants were cultured with vascular endothelial cells in the endothelial cell (EC) group, and implants were cultured without vascular endothelial cells in the blank group. Characterization of the cells was performed with immunofluorescence staining and a Transwell migration and cell tube formation assay. The levels of interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF) in the rabbit conjunctiva were measured with an ELISA. The results showed that the levels of IL-8 were decreased in the EC group and increased in the blank group. The levels of VEGF were increased in the EC group when compared to the blank group with statistical significance. The average depth of the fibrovascular tissue was obviously thicker in the EC group compared with that found in the blank group. These findings suggest that the vascular endothelial cell-hydroxyapatite orbital implant complex may be an effective strategy with which to accelerate vascularization and reduce complications of infection with satisfactory biological activity.
Collapse
Affiliation(s)
- Weiqi Wu
- Department of Ocular Oncology and Ocular Trauma, Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Sciences, Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China.,Department of Ophthalmology, The Third Affiliated Hospital of Nanchang University, Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Nanchang, Jiangxi 330008, P.R. China
| | - Hao Luo
- Department of Ocular Oncology and Ocular Trauma, Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Sciences, Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Dan Wu
- Department of Internal Medical, Nanchang HongDu Hospital of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, P.R. China
| | - Menglin Shi
- Department of Ocular Oncology and Ocular Trauma, Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Sciences, Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Jinhai Yu
- Department of Ocular Oncology and Ocular Trauma, Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Sciences, Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Hongfei Liao
- Department of Ocular Oncology and Ocular Trauma, Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Sciences, Key Laboratory of Ophthalmology of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
42
|
Hosseini M, Brown J, Shafiee A. Strategies to Induce Blood Vessel Ingrowth into Skin Grafts and Tissue-Engineered Substitutes. Tissue Eng Part C Methods 2022; 28:113-126. [PMID: 35172639 DOI: 10.1089/ten.tec.2021.0213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Skin is a multilayer organ consisting of several tissues and appendages residing in a complex niche. Adequate and physiologically regulated vascularization is an absolute requirement for skin homeostasis, regeneration, and wound healing. The lack of vascular networks and ischemia results in delayed wound closure. In addition, vascularization is critical for the prolonged function and survival of skin grafts and tissue-engineered skin substitutes. This study highlights the clinical challenges associated with the limited vascularization in the cutaneous wounds. Then, we highlight the novel approaches for the development of vascular networks in the skin autografts, allografts, and artificial substitutes. Also, the future directions to overcome the existing vascularization complications in skin grafting and synthetic skin substitutes are presented. Statement of Significance Delayed closure of large dermal wounds, such as burn injuries, results from the lack of vascular networks and ischemia. The amount of blood supply in the skin graft is the primary factor determining the quality of the transplanted grafts. The current skin grafts and their fabrication methods lack the appropriate features that contribute to the vascularization and integration of the wound bed and graft and adherence to the skin layers. Therefore, the new generation of skin grafts should consider advanced technologies to induce vascularization and overcome current challenges.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
| | - Jason Brown
- Herston Biofabrication Institute and Metro North Hospital and Health Service, Brisbane, Australia.,Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute and Metro North Hospital and Health Service, Brisbane, Australia.,Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, Australia.,UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
43
|
Wang Y, Kankala RK, Ou C, Chen A, Yang Z. Advances in hydrogel-based vascularized tissues for tissue repair and drug screening. Bioact Mater 2022; 9:198-220. [PMID: 34820566 PMCID: PMC8586021 DOI: 10.1016/j.bioactmat.2021.07.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The construction of biomimetic vasculatures within the artificial tissue models or organs is highly required for conveying nutrients, oxygen, and waste products, for improving the survival of engineered tissues in vitro. In recent times, the remarkable progress in utilizing hydrogels and understanding vascular biology have enabled the creation of three-dimensional (3D) tissues and organs composed of highly complex vascular systems. In this review, we give an emphasis on the utilization of hydrogels and their advantages in the vascularization of tissues. Initially, the significance of vascular elements and the regeneration mechanisms of vascularization, including angiogenesis and vasculogenesis, are briefly introduced. Further, we highlight the importance and advantages of hydrogels as artificial microenvironments in fabricating vascularized tissues or organs, in terms of tunable physical properties, high similarity in physiological environments, and alternative shaping mechanisms, among others. Furthermore, we discuss the utilization of such hydrogels-based vascularized tissues in various applications, including tissue regeneration, drug screening, and organ-on-chips. Finally, we put forward the key challenges, including multifunctionalities of hydrogels, selection of suitable cell phenotype, sophisticated engineering techniques, and clinical translation behind the development of the tissues with complex vasculatures towards their future development.
Collapse
Affiliation(s)
- Ying Wang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, PR China
| | - Caiwen Ou
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, PR China
| | - Zhilu Yang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| |
Collapse
|
44
|
Wu L, Tian K, Ding Z, Zhao T, Zhang X, Cheng W, Gao S, Lu Q, Kaplan DL. MSC‐Laden Composite Hydrogels for Inflammation and Angiogenic Regulation for Skin Flap Repair. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Lijun Wu
- Department of Plastic and Cosmetic Surgery The Second Affiliated Hospital of Soochow University Soochow University Suzhou 215004 P. R. China
| | - Kai Tian
- Department of Plastic and Cosmetic Surgery The Second Affiliated Hospital of Soochow University Soochow University Suzhou 215004 P. R. China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 P. R. China
| | - Tianlan Zhao
- Department of Plastic and Cosmetic Surgery The Second Affiliated Hospital of Soochow University Soochow University Suzhou 215004 P. R. China
| | - Xiaoyi Zhang
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 P. R. China
| | - Weinan Cheng
- Department of Orthopedics School of Medicine The First Affiliated Hospital of Xiamen University Xiamen University Xiamen 361000 P. R. China
| | - Suyue Gao
- Department of Dermatology and Cosmetic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou 215002 P. R. China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 P. R. China
| | - David L. Kaplan
- Department of Biomedical Engineering Tufts University Medford MA 02155 USA
| |
Collapse
|
45
|
Effect of Stromal Vascular Fractions on Angiogenesis of Injected Diced Cartilage. J Craniofac Surg 2022; 33:713-718. [PMID: 35013075 PMCID: PMC8865209 DOI: 10.1097/scs.0000000000007996] [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] [Indexed: 12/02/2022] Open
Abstract
Objectives: This study explored the effect of adipose-derived stromal vascular fractions (SVFs) on angiogenesis in injected autologous diced cartilage. Methods: Stromal vascular fractions were extracted by enzymatic digestion. Cartilage grafts were harvested from 1 side of the auricular cartilage of New Zealand rabbit and then diced to a size of 1.0 mm3. The grafts were divided into 2 groups. The control group was diced cartilage mixed with culture medium, and the experimental group was diced cartilage mixed with SVFs. The 2 groups of composite grafts were subcutaneously implanted on both sides of the back of each rabbit. After 4, 12 and 24 weeks, the tissue structure, number of blood vessels, and angiogenic factors in the grafts were observed. Results: The SVFs conformed to the current standard of the biological evaluation. Under an inverted microscope, the number of layers of chondrocytes in the experimental group was higher than that in the control group at 4 weeks. A small number of inflammatory cells and blood vessels were observed around the cartilage grafts. At 12 and 24 weeks, the volume of tissue was increased gradually by general observation. And a large number of chondrocytes were observed microscopically, whereas the number of inflammatory cells decreased. And meanwhile additional new blood vessels were observed. Immunohistochemical analysis of CD31 showed that the number of capillaries in the control group was significantly lower than that in the experimental group at 4, 12 and 24 weeks. Further, the expression of Hypoxia inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) mRNA and protein were measured by RT-PCR and Western bloting, respectively. The results showed that the mRNA expression of VEGF and HIF-1α in the experimental group was increased. The mRNA level remained higher than that of the control group at 24 weeks (P < 0.05). And the relative expression levels of VEGF and HIF-1α protein in the experimental group were higher than those in the control group at 4, 12 and 24 weeks (P < 0.05). Conclusion: Autologous diced cartilage mixed with adipose-derived SVFs can promote angiogenesis when transplanted by injection. Further research showed that SVFs could increase the expression levels of VEGF and HIF-1α in the grafts, which may be part of the mechanism that SVFs promoted the angiogenesis of diced cartilage.
Collapse
|
46
|
Yuan X, Li L, Liu H, Luo J, Zhao Y, Pan C, Zhang X, Chen Y, Gou M. Strategies for improving adipose-derived stem cells for tissue regeneration. BURNS & TRAUMA 2022; 10:tkac028. [PMID: 35992369 PMCID: PMC9382096 DOI: 10.1093/burnst/tkac028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/27/2022] [Indexed: 11/13/2022]
Abstract
Abstract
Adipose-derived stem cells (ADSCs) have promising applications in tissue regeneration. Currently, there are only a few ADSC products that have been approved for clinical use. The clinical application of ADSCs still faces many challenges. Here, we review emerging strategies to improve the therapeutic efficacy of ADSCs in tissue regeneration. First, a great quantity of cells is often needed for the stem cell therapies, which requires the advanced cell expansion technologies. In addition cell-derived products are also required for the development of ‘cell-free’ therapies to overcome the drawbacks of cell-based therapies. Second, it is necessary to strengthen the regenerative functions of ADSCs, including viability, differentiation and paracrine ability, for the tissue repair and regeneration required for different physiological and pathophysiological conditions. Third, poor delivery efficiency also restricts the therapeutic effect of ADSCs. Effective methods to improve cell delivery include alleviating harsh microenvironments, enhancing targeting ability and prolonging cell retention. Moreover, we also point out some critical issues about the sources, effectiveness and safety of ADSCs. With these advanced strategies to improve the therapeutic efficacy of ADSCs, ADSC-based treatment holds great promise for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Xin Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Li Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Haofan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Jing Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Yongchao Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Cheng Pan
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Xue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Yuwen Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| |
Collapse
|
47
|
Wong J, Murphy M, Wu YF, Murphy R, Frueh FS, Farnebo S. Basic science approaches to common hand surgery problems. J Hand Surg Eur Vol 2022; 47:117-126. [PMID: 34472390 DOI: 10.1177/17531934211042697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The field of hand surgery is constantly evolving to meet challenges of populations with increasing age and higher demands for active living. While our surgical care has improved over the last decades, it seems that future major improvement in outcomes of clinical treatment will come through advances in biologics and the translation of major discoveries in basic science. This article aims to provide an update on where basic science solutions may answer some of the most critical issues in hand surgery, with a focus on augmentation of tissue repair.
Collapse
Affiliation(s)
- Jason Wong
- Blond McIndoe Laboratories, Manchester, UK.,Department of Plastic Surgery, University of Manchester and Manchester University Foundation Trust, Manchester, UK
| | - Matthew Murphy
- Blond McIndoe Laboratories, Manchester, UK.,Department of Plastic Surgery, University of Manchester and Manchester University Foundation Trust, Manchester, UK
| | - Ya Fang Wu
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ralph Murphy
- Blond McIndoe Laboratories, Manchester, UK.,Department of Plastic Surgery, University of Manchester and Manchester University Foundation Trust, Manchester, UK
| | - Florian S Frueh
- Department of Plastic Surgery and Hand Surgery, University of Zurich, Zurich, Switzerland
| | - Simon Farnebo
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Plastic Surgery, Hand Surgery, and Burns, Linköping University, Linköping, Sweden
| |
Collapse
|
48
|
Spheroids of Bladder Smooth Muscle Cells for Bladder Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9391575. [PMID: 34805410 PMCID: PMC8601859 DOI: 10.1155/2021/9391575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/28/2021] [Indexed: 12/02/2022]
Abstract
Cell-based tissue engineering (TE) has been proposed to improve treatment outcomes in end-stage bladder disease, but TE approaches with 2D smooth muscle cell (SMC) culture have so far been unsuccessful. Here, we report the development of primary bladder-derived 3D SMC spheroids that outperform 2D SMC cultures in differentiation, maturation, and extracellular matrix (ECM) production. Bladder SMC spheroids were compared with 2D cultures using live-dead staining, qRT-PCR, immunofluorescence, and immunoblotting to investigate culture conditions, contractile phenotype, and ECM deposition. The SMC spheroids were viable for up to 14 days and differentiated rather than proliferating. Spheroids predominantly expressed the late myogenic differentiation marker MyH11, whereas 2D SMC expressed more of the general SMC differentiation marker α-SMA and less MyH11. Furthermore, the expression of bladder wall-specific ECM proteins in SMC spheroids was markedly higher. This first establishment and analysis of primary bladder SMC spheroids are particularly promising for TE because differentiated SMCs and ECM deposition are a prerequisite to building a functional bladder wall substitute. We were able to confirm that SMC spheroids are promising building blocks for studying detrusor regeneration in detail and may provide improved function and regenerative potential, contributing to taking bladder TE a significant step forward.
Collapse
|
49
|
Zhao Z, Sun Y, Qiao Q, Zhang L, Xie X, Weir MD, Schneider A, Xu HHK, Zhang N, Zhang K, Bai Y. Human Periodontal Ligament Stem Cell and Umbilical Vein Endothelial Cell Co-Culture to Prevascularize Scaffolds for Angiogenic and Osteogenic Tissue Engineering. Int J Mol Sci 2021; 22:ijms222212363. [PMID: 34830243 PMCID: PMC8621970 DOI: 10.3390/ijms222212363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background: Vascularization remains a critical challenge in bone tissue engineering. The objective of this study was to prevascularize calcium phosphate cement (CPC) scaffold by co-culturing human periodontal ligament stem cells (hPDLSCs) and human umbilical vein endothelial cells (hUVECs) for the first time; (2) Methods: hPDLSCs and/or hUVECs were seeded on CPC scaffolds. Three groups were tested: (i) hUVEC group (hUVECs on CPC); (ii) hPDLSC group (hPDLSCs on CPC); (iii) co-culture group (hPDLSCs + hUVECs on CPC). Osteogenic differentiation, bone mineral synthesis, and microcapillary-like structures were evaluated; (3) Results: Angiogenic gene expressions of co-culture group were 6–9 fold those of monoculture. vWF expression of co-culture group was 3 times lower than hUVEC-monoculture group. Osteogenic expressions of co-culture group were 2–3 folds those of the hPDLSC-monoculture group. ALP activity and bone mineral synthesis of co-culture were much higher than hPDLSC-monoculture group. Co-culture group formed capillary-like structures at 14–21 days. Vessel length and junction numbers increased with time; (4) Conclusions: The hUVECs + hPDLSCs co-culture on CPC scaffold achieved excellent osteogenic and angiogenic capability in vitro for the first time, generating prevascularized networks. The hPDLSCs + hUVECs co-culture had much better osteogenesis and angiogenesis than monoculture. CPC scaffolds prevacularized via hPDLSCs + hUVECs are promising for dental, craniofacial, and orthopedic applications.
Collapse
Affiliation(s)
- Zeqing Zhao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
| | - Yaxi Sun
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
| | - Li Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
| | - Xianju Xie
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
| | - Michael D. Weir
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; (M.D.W.); (H.H.K.X.)
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA;
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hockin H. K. Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; (M.D.W.); (H.H.K.X.)
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ning Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
- Correspondence: (N.Z.); (Y.B.)
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China; (Z.Z.); (Y.S.); (Q.Q.); (L.Z.); (X.X.); (K.Z.)
- Correspondence: (N.Z.); (Y.B.)
| |
Collapse
|
50
|
Nguyen N, Thurgood P, Sekar NC, Chen S, Pirogova E, Peter K, Baratchi S, Khoshmanesh K. Microfluidic models of the human circulatory system: versatile platforms for exploring mechanobiology and disease modeling. Biophys Rev 2021; 13:769-786. [PMID: 34777617 DOI: 10.1007/s12551-021-00815-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human circulatory system is a marvelous fluidic system, which is very sensitive to biophysical and biochemical cues. The current animal and cell culture models do not recapitulate the functional properties of the human circulatory system, limiting our ability to fully understand the complex biological processes underlying the dysfunction of this multifaceted system. In this review, we discuss the unique ability of microfluidic systems to recapitulate the biophysical, biochemical, and functional properties of the human circulatory system. We also describe the remarkable capacity of microfluidic technologies for exploring the complex mechanobiology of the cardiovascular system, mechanistic studying of cardiovascular diseases, and screening cardiovascular drugs with the additional benefit of reducing the need for animal models. We also discuss opportunities for further advancement in this exciting field.
Collapse
Affiliation(s)
- Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Sheng Chen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiometabolic Health, The University of Melbourne, Parkville, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | | |
Collapse
|