1
|
Hromada C, Heimel P, Kerbl M, Gál L, Nürnberger S, Schaedl B, Ferguson J, Swiadek N, Monforte X, Heinzel JC, Nógrádi A, Teuschl-Woller AH, Hercher D. Silk-based nerve guidance conduits with macroscopic holes modulate the vascularization of regenerating rat sciatic nerve. Neural Regen Res 2025; 20:1789-1800. [PMID: 39104116 PMCID: PMC11688571 DOI: 10.4103/nrr.nrr-d-23-01518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/18/2023] [Accepted: 02/01/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00029/figure1/v/2024-08-05T133530Z/r/image-tiff Peripheral nerve injuries induce a severe motor and sensory deficit. Since the availability of autologous nerve transplants for nerve repair is very limited, alternative treatment strategies are sought, including the use of tubular nerve guidance conduits (tNGCs). However, the use of tNGCs results in poor functional recovery and central necrosis of the regenerating tissue, which limits their application to short nerve lesion defects (typically shorter than 3 cm). Given the importance of vascularization in nerve regeneration, we hypothesized that enabling the growth of blood vessels from the surrounding tissue into the regenerating nerve within the tNGC would help eliminate necrotic processes and lead to improved regeneration. In this study, we reported the application of macroscopic holes into the tubular walls of silk-based tNGCs and compared the various features of these improved silk+ tNGCs with the tubes without holes (silk- tNGCs) and autologous nerve transplants in an 8-mm sciatic nerve defect in rats. Using a combination of micro-computed tomography and histological analyses, we were able to prove that the use of silk+ tNGCs induced the growth of blood vessels from the adjacent tissue to the intraluminal neovascular formation. A significantly higher number of blood vessels in the silk+ group was found compared with autologous nerve transplants and silk-, accompanied by improved axon regeneration at the distal coaptation point compared with the silk- tNGCs at 7 weeks postoperatively. In the 15-mm (critical size) sciatic nerve defect model, we again observed a distinct ingrowth of blood vessels through the tubular walls of silk+ tNGCs, but without improved functional recovery at 12 weeks postoperatively. Our data proves that macroporous tNGCs increase the vascular supply of regenerating nerves and facilitate improved axonal regeneration in a short-defect model but not in a critical-size defect model. This study suggests that further optimization of the macroscopic holes silk+ tNGC approach containing macroscopic holes might result in improved grafting technology suitable for future clinical use.
Collapse
Affiliation(s)
- Carina Hromada
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Patrick Heimel
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Markus Kerbl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Landesklinikum Wiener Neustadt, 2700 Wiener Neustadt, Austria
| | - László Gál
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Sylvia Nürnberger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- Medical University of Vienna, Department of Orthopedics and Trauma Surgery, Devision of Trauma Surgery, Vienna, Austria
| | - Barbara Schaedl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - James Ferguson
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Nicole Swiadek
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Xavier Monforte
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Johannes C. Heinzel
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Unfallklinik Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Andreas H. Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - David Hercher
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| |
Collapse
|
2
|
Han Y, Liu Z, Song C. Fenugreek seed extract combined with acellular nerve allografts promotes peripheral nerve regeneration and neovascularization in sciatic nerve defects. Regen Ther 2025; 28:383-393. [PMID: 39896442 PMCID: PMC11787413 DOI: 10.1016/j.reth.2024.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/02/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Background Acellular nerve allografts (ANAs) have been confirmed to improve the repair and reconstruction of long peripheral nerve defects. However, its efficacy is not comparable to that of autologous nerve grafts, which are used as the gold standard for treating peripheral nerve defects. Our study investigated whether fenugreek seed extract (FSE) exhibits neuroprotective potential and enhances the therapeutic outcomes of ANA repair in peripheral nerve defects. Methods Rat Schwann cells were treated with FSE to assess the effects of FSE on cell proliferation and their secretion function of neurotrophic factors in vitro. Sprague-Dawley rats with a unilateral 15-mm sciatic nerve defect were randomized into the ANA group (the 15-mm defect was replaced by an 18-mm ANA), the ANA + FSE group (the 15-mm defect was repaired with an 18-mm ANA with FSE administration for four weeks), and the Auto group (the 15-mm defect was repaired with an autologous graft). After four weeks post-surgery, various behavioral tests and electrophysiological assays were performed to evaluate the motor and sensory behavior as well as nerve conduction of rats. Then, rats were sacrificed, and the nerve grafts were collected for toluidine blue staining, RT-qPCR, immunofluorescence staining, immunohistochemical staining to evaluate nerve regeneration, neovascularization, and neuroinflammation. Their gastrocnemius was harvested for Masson's trichrome staining to examine gastrocnemius muscle recovery. Results FSE treatment promoted Schwann cell proliferation and its secretion of neurotrophic factors in vitro. Compared with ANAs alone, FSE treatment combined with ANAs enhanced axonal regeneration, upregulated S100, NF200, P0, MBP, and GAP43 expression, facilitated angiogenesis, and elevated neurotrophic factor expression in regenerating nerves of rats with sciatic nerve defects. In addition, FSE treatment promoted gastrocnemius muscle recovery, stimulated motor and sensory functional recovery and nerve conduction, and mitigated neuroinflammation in rats with sciatic nerve defects after repair with ANAs. Conclusion FSE treatment improves the beneficial effects of ANA repair on sciatic nerve defects.
Collapse
Affiliation(s)
- Yuanyuan Han
- Department of Neurology, Suqian First Hospital, Suzhi Road 120, Suqian, 223800 Jiangsu Province, China
| | - Zhiwei Liu
- Department of Neurology, Suqian First Hospital, Suzhi Road 120, Suqian, 223800 Jiangsu Province, China
| | - Chunjie Song
- Department of Neurology, Suqian First Hospital, Suzhi Road 120, Suqian, 223800 Jiangsu Province, China
| |
Collapse
|
3
|
Shen M, Ye X, Zhou Q, Zheng M, Du M, Wang L, Cong M, Liu C, Deng C, Xu Z, Wang Y, Li J, Feng M, Ye Y, Zhang S, Xu W, Lu Y, Kong J, Gong J, Xia Y, Gu J, Xie H, He Q, Zhang Q, Sun H, Liu X, Gong L, Yu M, Gu X, Zhao J, Zhang N, Ding F, Zhou S. Angiogenesis-promoting effect of SKP-SC-EVs-derived miRNA-30a-5p in peripheral nerve regeneration by targeting LIF and ANGPT2. J Biol Chem 2024; 301:108146. [PMID: 39732166 PMCID: PMC11791313 DOI: 10.1016/j.jbc.2024.108146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 11/24/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
Ischemia and hypoxia caused by vascular injury intensify nerve damage. Skin precursor-derived Schwann cells have demonstrated an accelerated in vivo prevascularization of tissue-engineered nerves. Furthermore, extracellular vesicles from skin precursor-derived Schwann cells (SKP-SC-EVs) show the potential in aiding peripheral nerve regeneration. Nonetheless, the capacity of SKP-SC-EVs to facilitate nerve repair via angiogenesis remains uncertain. This study observed that SKP-SC-EVs significantly enhanced angiogenesis, evidenced by increased transparency of the tissue-engineered nerve graft and ultrasonic blood flow imaging. In vitro experiments confirmed that SKP-SC-EVs promote the proliferation, migration, and tube formation of human umbilical vein endothelial cells, a standard model for assessing angiogenic potential. Additionally, a comprehensive miRNA expression profile of SKP-SC-EVs was performed, leading to the identification of potential candidates through functional experiments. Among these, miR-30a-5p emerged as a significant candidate, demonstrating remarkable proangiogenic effects both in vivo and in vitro, akin to the effects of SKP-SC-EVs. Furthermore, luciferase reporter assay and functional experiments revealed that miR-30a-5p in SKP-SC-EVs promotes angiogenesis by targeting ANGPT2 and LIF without sufficient VEGFa. Thus, the enrichment of miR-30a-5p in SKP-SC-EVs indicates its pivotal role as a regulator of angiogenesis, presenting a promising avenue for cell-free treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Xinli Ye
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Qiang Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Mingzhi Du
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Lijuan Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Meng Cong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Chunyan Deng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Zhen Xu
- Department of Clinical Medical Research Center, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu Province, China
| | - Yu Wang
- Department of Clinical Medical Research Center, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu Province, China
| | - Jiyu Li
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Min Feng
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yujiao Ye
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shuyu Zhang
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Wenqing Xu
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yi Lu
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Junjie Kong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Jiahuan Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Yingjie Xia
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jinhua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong, China
| | - Huimin Xie
- The Affiliated Nantong Stomatological Hospital of Nantong University, Nantong, China
| | - Qianru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Qi Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Xingjun Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Miaomei Yu
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Jian Zhao
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Ning Zhang
- Department of Clinical Medical Research Center, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu Province, China; Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China; Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
4
|
Muangsanit P, Smith P. Harnessing endothelial cells and vascularization strategies for nerve regeneration. Neural Regen Res 2024; 19:2337-2338. [PMID: 38526263 PMCID: PMC11090433 DOI: 10.4103/nrr.nrr-d-23-01840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/25/2023] [Accepted: 01/05/2024] [Indexed: 03/26/2024] Open
Affiliation(s)
- Papon Muangsanit
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Poppy Smith
- UCL Centre for Nerve Engineering, UCL, London, UK; Department of Pharmacology, UCL School of Pharmacy, UCL, London, UK
| |
Collapse
|
5
|
Boyce L, Wormald JCR, Ng CY, Miller R. A systematic review of clinical and laboratory studies comparing vascularised versus non-vascularised nerve grafts in peripheral nerve reconstruction. J Plast Reconstr Aesthet Surg 2024; 97:182-199. [PMID: 39168029 DOI: 10.1016/j.bjps.2024.07.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/11/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Peripheral nerve injuries (PNIs) are common, with complex defects posing a significant reconstructive challenge. Although vascularised (VNGs) and non-vascularised nerve grafts (NVNGs) are established treatment options, there is no comprehensive summary of the evidence supporting their clinical, electrophysiological, and histological outcomes. This review aims to systematically evaluate the clinical and laboratory literature comparing VNGs and NVNGs to inform future clinical practice and research. METHODS This review was prospectively registered and reported according to PRISMA guidelines. PubMed, EMBASE, SCOPUS, and the Cochrane Register were systematically searched. Studies comparing VNGs and NVNGs in PNIs were included. Meta-analyses were performed for outcomes reported in ≥3 laboratory studies. Functional outcomes were synthesised by vote-counting based on direction of effect for clinical studies. Risk-of-bias was assessed using RoB2, ROBINS-I, and SYRCLE, and the certainty of evidence was evaluated using GRADE. RESULTS Seven clinical and 34 laboratory studies were included. Of the clinical comparisons, 90% and 56% identified an effect on recovery of sensibility (p = 0.01) and motor function (p = 0.05), respectively, that favoured VNGs. Nine (of 13) separate meta-analyses of laboratory studies demonstrated reduced muscular atrophy, superior axonal regeneration, and remyelination in VNGs. VNGs eliminated the 3-day interval of ischaemia otherwise sustained by NVNGs. Overall, the quality of evidence was low. CONCLUSION This systematic review indicates that VNGs may offer some advantages over NVNGs in PNI reconstruction. However, due to the low quality of evidence, significant statistical heterogeneity, and clinical diversity of the included studies, these conclusions should be interpreted with caution. Further high-quality clinical trials are necessary to validate these findings.
Collapse
Affiliation(s)
| | - Justin Conrad Rosen Wormald
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK.
| | | | | |
Collapse
|
6
|
Rasouli A, Roshangar L, Hosseini M, Pourmohammadfazel A, Nikzad S. Beyond boundaries: The therapeutic potential of exosomes in neural microenvironments in neurological disorders. Neuroscience 2024; 553:98-109. [PMID: 38964450 DOI: 10.1016/j.neuroscience.2024.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Neurological disorders are a diverse group of conditions that can significantly impact individuals' quality of life. The maintenance of neural microenvironment homeostasis is essential for optimal physiological cellular processes. Perturbations in this delicate balance underlie various pathological manifestations observed across various neurological disorders. Current treatments for neurological disorders face substantial challenges, primarily due to the formidable blood-brain barrier and the intricate nature of neural tissue structures. These obstacles have resulted in a paucity of effective therapies and inefficiencies in patient care. Exosomes, nanoscale vesicles that contain a complex repertoire of biomolecules, are identifiable in various bodily fluids. They hold substantial promise in numerous therapeutic interventions due to their unique attributes, including targeted drug delivery mechanisms and the ability to cross the BBB, thereby enhancing their therapeutic potential. In this review, we investigate the therapeutic potential of exosomes across a range of neurological disorders, including neurodegenerative disorders, traumatic brain injury, peripheral nerve injury, brain tumors, and stroke. Through both in vitro and in vivo studies, our findings underscore the beneficial influence of exosomes in enhancing the neural microenvironment following neurological diseases, offering promise for improved neural recovery and management in these conditions.
Collapse
Affiliation(s)
- Arefe Rasouli
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammadbagher Hosseini
- Department of Pediatrics, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Pourmohammadfazel
- Department of Anatomical Sciences, School of Medicine Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
7
|
Patel D, Shetty S, Acha C, Pantoja IEM, Zhao A, George D, Gracias DH. Microinstrumentation for Brain Organoids. Adv Healthc Mater 2024; 13:e2302456. [PMID: 38217546 DOI: 10.1002/adhm.202302456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/10/2023] [Indexed: 01/15/2024]
Abstract
Brain organoids are three-dimensional aggregates of self-organized differentiated stem cells that mimic the structure and function of human brain regions. Organoids bridge the gaps between conventional drug screening models such as planar mammalian cell culture, animal studies, and clinical trials. They can revolutionize the fields of developmental biology, neuroscience, toxicology, and computer engineering. Conventional microinstrumentation for conventional cellular engineering, such as planar microfluidic chips; microelectrode arrays (MEAs); and optical, magnetic, and acoustic techniques, has limitations when applied to three-dimensional (3D) organoids, primarily due to their limits with inherently two-dimensional geometry and interfacing. Hence, there is an urgent need to develop new instrumentation compatible with live cell culture techniques and with scalable 3D formats relevant to organoids. This review discusses conventional planar approaches and emerging 3D microinstrumentation necessary for advanced organoid-machine interfaces. Specifically, this article surveys recently developed microinstrumentation, including 3D printed and curved microfluidics, 3D and fast-scan optical techniques, buckling and self-folding MEAs, 3D interfaces for electrochemical measurements, and 3D spatially controllable magnetic and acoustic technologies relevant to two-way information transfer with brain organoids. This article highlights key challenges that must be addressed for robust organoid culture and reliable 3D spatiotemporal information transfer.
Collapse
Affiliation(s)
- Devan Patel
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Saniya Shetty
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Chris Acha
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Itzy E Morales Pantoja
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Alice Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Derosh George
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - David H Gracias
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD, 21218, USA
- Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Center for MicroPhysiological Systems (MPS), Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
8
|
Wu RT, Chuieng-Yi Lu J. Nerve Autograft: Preservation of a Lost Art. Hand Clin 2024; 40:347-356. [PMID: 38972679 DOI: 10.1016/j.hcl.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Nerve autografts involve the transplantation of a segment of the patient's own nerve to bridge a nerve gap. Autografts provide biological compatibility, support for axonal regeneration, and the ability to provide an anatomic scaffold for regrowth that other modalities may not match. Disadvantages of the autograft include donor site morbidity and the extra operative time needed to harvest the graft. Nevertheless, nerve autografts such as the sural nerve remain the gold standard in reconstructing nerve gaps, but a multitude of factors need to be favorable in order to garner reliable, consistent outcomes.
Collapse
Affiliation(s)
- Robin T Wu
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Standford, CA 94305, USA
| | - Johnny Chuieng-Yi Lu
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, No. 5, Fuxing Street, Guishan District, Taoyuan City, 333 Taiwan; Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
9
|
Ramasubbu K, Venkatraman G, Ramanathan G, Dhanasekar S, Rajeswari VD. Molecular and cellular signalling pathways for promoting neural tissue growth - A tissue engineering approach. Life Sci 2024; 346:122640. [PMID: 38614302 DOI: 10.1016/j.lfs.2024.122640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neural tissue engineering is a sub-field of tissue engineering that develops neural tissue. Damaged central and peripheral nervous tissue can be fabricated with a suitable scaffold printed with biomaterials. These scaffolds promote cell growth, development, and migration, yet they vary according to the biomaterial and scaffold printing technique, which determine the physical and biochemical properties. The physical and biochemical properties of scaffolds stimulate diverse signalling pathways, such as Wnt, NOTCH, Hedgehog, and ion channels- mediated pathways to promote neuron migration, elongation and migration. However, neurotransmitters like dopamine, acetylcholine, gamma amino butyric acid, and other signalling molecules are critical in neural tissue engineering to tissue fabrication. Thus, this review focuses on neural tissue regeneration with a tissue engineering approach highlighting the signalling pathways. Further, it explores the interaction of the scaffolds with the signalling pathways for generating neural tissue.
Collapse
Affiliation(s)
- Kanagavalli Ramasubbu
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Ganasambanthan Ramanathan
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Sivaraman Dhanasekar
- Department of Biotechnology, Pandit Deendayal Energy University, Gandhinagar 382007, Gujarat, India
| | - V Devi Rajeswari
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
10
|
Quizon MJ, Deppen JN, Barber GF, Kalelkar PP, Coronel MM, Levit RD, García AJ. VEGF-delivering PEG hydrogels promote vascularization in the porcine subcutaneous space. J Biomed Mater Res A 2024; 112:866-880. [PMID: 38189109 PMCID: PMC10984793 DOI: 10.1002/jbm.a.37666] [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: 11/16/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024]
Abstract
For cell therapies, the subcutaneous space is an attractive transplant site due to its large surface area and accessibility for implantation, monitoring, biopsy, and retrieval. However, its poor vascularization has catalyzed research to induce blood vessel formation within the site to enhance cell revascularization and survival. Most studies focus on the subcutaneous space of rodents, which does not recapitulate important anatomical features and vascularization responses of humans. Herein, we evaluate biomaterial-driven vascularization in the porcine subcutaneous space. Additionally, we report the first use of cost-effective fluorescent microspheres to quantify perfusion in the porcine subcutaneous space. We investigate the vascularization-inducing efficacy of vascular endothelial growth factor (VEGF)-delivering synthetic hydrogels based on 4-arm poly(ethylene) glycol macromers with terminal maleimides (PEG-4MAL). We compare three groups: a non-degradable hydrogel with a VEGF-releasing PEG-4MAL gel coating (Core+VEGF gel); an uncoated, non-degradable hydrogel (Core-only); and naïve tissue. After 2 weeks, Core+VEGF gel has significantly higher tissue perfusion, blood vessel area, blood vessel density, and number of vessels compared to both Core-only and naïve tissue. Furthermore, healthy vital signs during surgery and post-procedure metrics demonstrate the safety of hydrogel delivery. We demonstrate that VEGF-delivering synthetic hydrogels induce robust vascularization and perfusion in the porcine subcutaneous space.
Collapse
Affiliation(s)
- Michelle J. Quizon
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Juline N. Deppen
- Division of Cardiology, Emory University School of Medicine, 1440 Clifton Rd, Atlanta, GA 30322, USA
| | - Graham F. Barber
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Pranav P. Kalelkar
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - María M. Coronel
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Rebecca D. Levit
- Division of Cardiology, Emory University School of Medicine, 1440 Clifton Rd, Atlanta, GA 30322, USA
| | - Andrés J. García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| |
Collapse
|
11
|
Liu L, Chen M, Zhang J, Li H, Li Z, Song J, Ma S, Wang Y, Lou X. Oriented polyaniline/poly-l-lactic acid/gelatin nanofiber scaffolds promote outgrowth of spiral ganglion neurons. J Biomed Mater Res A 2024; 112:700-709. [PMID: 37962013 DOI: 10.1002/jbm.a.37649] [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: 07/21/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023]
Abstract
Sensorineural hearing loss (SNHL) is caused by the loss of sensory hair cells (HCs) and/or connected spiral ganglion neurons (SGNs). The current clinical conventional treatment for SNHL is cochlear implantation (CI). The principle of CI is to bypass degenerated auditory HCs and directly electrically stimulate SGNs to restore hearing. However, the effectiveness of CI is limited when SGNs are severely damaged. In the present study, oriented nanofiber scaffolds were fabricated using electrospinning technology to mimic the SGN spatial microenvironment in the inner ear. Meanwhile, different proportions of polyaniline (PANI), poly-l-lactide (PLLA), gelatin (Gel) were composited to mimic the composition and mechanical properties of auditory basement membrane. The effects of oriented PANI/PLLA/Gel biomimetic nanofiber scaffolds for neurite outgrowth were analyzed. The results showed the SGNs grew in an orientation along the fiber direction, and the length of the protrusions increased significantly on PANI/PLLA/Gel scaffold groups. The 2% PANI/PLLA/Gel group showed best effects for promoting SGN adhesion and nerve fiber extension. In conclusion, the biomimetic oriented nanofiber scaffolds can simulate the microenvironment of SGNs as well as promote neurite outgrowth in vitro, which may provide a feasible research idea for SGN regeneration and even therapeutic treatments of SNHL in future.
Collapse
Affiliation(s)
- Li Liu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Mengyu Chen
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Junming Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Haobo Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Zhaoxia Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Jianhao Song
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Shutao Ma
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Yingjie Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Xiangxin Lou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| |
Collapse
|
12
|
Cheng Q, Wang W, Dong X, Chai Y, Goto T, Tu R, Yan L, Yu A, Dai H. An Adaptable Drug Delivery System Facilitates Peripheral Nerve Repair by Remodeling the Microenvironment. Biomacromolecules 2024; 25:1509-1526. [PMID: 38376392 DOI: 10.1021/acs.biomac.3c01094] [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: 02/21/2024]
Abstract
The multifaceted process of nerve regeneration following damage remains a significant clinical issue, due to the lack of a favorable regenerative microenvironment and insufficient endogenous biochemical signaling. However, the current nerve grafts have limitations in functionality, as they require a greater capacity to effectively regulate the intricate microenvironment associated with nerve regeneration. In this regard, we proposed the construction of a functional artificial scaffold based on a "two-pronged" approach. The whole system was developed by encapsulating Tazarotene within nanomicelles formed through self-assembly of reactive oxygen species (ROS)-responsive amphiphilic triblock copolymer, all of which were further loaded into a thermosensitive injectable hydrogel. Notably, the hydrogel exhibits obvious temperature sensitivity at a concentration of 6 wt %, and the nanoparticles possess concentration-dependent H2O2-response capability with a controlled release profile in 48 h. The combined strategy promoted the repair of injured peripheral nerves, attributed to the dual role of the materials, which mainly involved providing structural support, modulating the immune microenvironment, and enhancing angiogenesis. Overall, this study opens up intriguing prospects in tissue engineering.
Collapse
Affiliation(s)
- Qiang Cheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Weixing Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xianzhen Dong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Yunhui Chai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Takashi Goto
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Rong Tu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan 528200, China
| |
Collapse
|
13
|
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
|
14
|
Giglia G, Rosatti F, Giannone AG, Gambino G, Zizzo MG, Florena AM, Sardo P, Toia F. Vascularized versus Free Nerve Grafts: An Experimental Study on Rats. J Pers Med 2023; 13:1682. [PMID: 38138909 PMCID: PMC10744603 DOI: 10.3390/jpm13121682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/13/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Vascularized nerve grafts (VNGs) have been proposed as a superior alternative to free nerve grafts (FNGs) for complex nerve defects. A greater regenerative potential has been suggested by clinical and experimental studies, but conclusive evidence is still lacking. METHODS In this experimental study, 10 adult male Wistar rats received a non-vascularized orthotopic sciatic nerve graft on their right side, and a vascularized orthotopic sciatic nerve graft nerve on their left side. Functional outcome following nerve regeneration was evaluated through electrodiagnostic studies, target muscles weight and histomorphology, and data of VNGs and FNGs were compared. RESULTS The results of this study showed a significant difference in the motor unit number of Gastrocnemius Medialis (GM) estimated by MUNE in the VNG side compared to the FNG side. No other significant differences in axonal regeneration and muscle reinnervation were evident at either electrodiagnostic, histomorphology studies or muscle weight. CONCLUSIONS This experimental model showed slight differences in nerve regeneration between VNGs and FNGs, but cannot support a high clinical advantage for VNGs. The results of this study show that VNGs are not strongly superior to FNGs in the rat model, even in avascular beds. Clinical advantages of VNGs are likely to be limited to extensive and thick nerve defects and can only be assessed on experimental model with bigger animals. Also, we showed that the MUNE technique provided a reliable and reproducible evaluation of functional outcomes in the rat sciatic nerve and defined a reproducible protocol for functional evaluation of muscle reinnervation.
Collapse
Affiliation(s)
- Giuseppe Giglia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (G.G.); (G.G.); (P.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Fernando Rosatti
- Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy;
| | - Antonino Giulio Giannone
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.G.G.); (A.M.F.)
| | - Giuditta Gambino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (G.G.); (G.G.); (P.S.)
| | - Maria Grazia Zizzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90127 Palermo, Italy;
- ATeN (Advanced Technologies Network) Center, University of Palermo, Viale delle Scienze, 90127 Palermo, Italy
| | - Ada Maria Florena
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.G.G.); (A.M.F.)
| | - Pierangelo Sardo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90127 Palermo, Italy; (G.G.); (G.G.); (P.S.)
| | - Francesca Toia
- Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
15
|
Hu J, Jiang Z, Zhang J, Yang G. Application of silk fibroin coatings for biomaterial surface modification: a silk road for biomedicine. J Zhejiang Univ Sci B 2023; 24:943-956. [PMID: 37961798 PMCID: PMC10646393 DOI: 10.1631/jzus.b2300003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/18/2023] [Indexed: 11/15/2023]
Abstract
Silk fibroin (SF) as a natural biopolymer has become a popular material for biomedical applications due to its minimal immunogenicity, tunable biodegradability, and high biocompatibility. Nowadays, various techniques have been developed for the applications of SF in bioengineering. Most of the literature reviews focus on the SF-based biomaterials and their different forms of applications such as films, hydrogels, and scaffolds. SF is also valuable as a coating on other substrate materials for biomedicine; however, there are few reviews related to SF-coated biomaterials. Thus, in this review, we focused on the surface modification of biomaterials using SF coatings, demonstrated their various preparation methods on substrate materials, and introduced the latest procedures. The diverse applications of SF coatings for biomedicine are discussed, including bone, ligament, skin, mucosa, and nerve regeneration, and dental implant surface modification. SF coating is conducive to inducing cell adhesion and migration, promoting hydroxyapatite (HA) deposition and matrix mineralization, and inhibiting the Notch signaling pathway, making it a promising strategy for bone regeneration. In addition, SF-coated composite scaffolds can be considered prospective candidates for ligament regeneration after injury. SF coating has been proven to enhance the mechanical properties of the substrate material, and render integral stability to the dressing material during the regeneration of skin and mucosa. Moreover, SF coating is a potential strategy to accelerate nerve regeneration due to its dielectric properties, mechanical flexibility, and angiogenesis promotion effect. In addition, SF coating is an effective and popular means for dental implant surface modification to promote osteogenesis around implants made of different materials. Thus, this review can be of great benefit for further improvements in SF-coated biomaterials, and will undoubtedly contribute to clinical transformation in the future.
Collapse
Affiliation(s)
- Jinxing Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
16
|
Namini MS, Daneshimehr F, Beheshtizadeh N, Mansouri V, Ai J, Jahromi HK, Ebrahimi-Barough S. Cell-free therapy based on extracellular vesicles: a promising therapeutic strategy for peripheral nerve injury. Stem Cell Res Ther 2023; 14:254. [PMID: 37726794 PMCID: PMC10510237 DOI: 10.1186/s13287-023-03467-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023] Open
Abstract
Peripheral nerve injury (PNI) is one of the public health concerns that can result in a loss of sensory or motor function in the areas in which injured and non-injured nerves come together. Up until now, there has been no optimized therapy for complete nerve regeneration after PNI. Exosome-based therapies are an emerging and effective therapeutic strategy for promoting nerve regeneration and functional recovery. Exosomes, as natural extracellular vesicles, contain bioactive molecules for intracellular communications and nervous tissue function, which could overcome the challenges of cell-based therapies. Furthermore, the bioactivity and ability of exosomes to deliver various types of agents, such as proteins and microRNA, have made exosomes a potential approach for neurotherapeutics. However, the type of cell origin, dosage, and targeted delivery of exosomes still pose challenges for the clinical translation of exosome therapeutics. In this review, we have focused on Schwann cell and mesenchymal stem cell (MSC)-derived exosomes in nerve tissue regeneration. Also, we expressed the current understanding of MSC-derived exosomes related to nerve regeneration and provided insights for developing a cell-free MSC therapeutic strategy for nerve injury.
Collapse
Affiliation(s)
- Mojdeh Salehi Namini
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Daneshimehr
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Kargar Jahromi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
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
|
18
|
Matzkeit N, Kisch T, Waldmann A, Schweiger U, Mailänder P, Westermair AL. Deep wrist injuries from suicide attempts vs. accidents do not differ regarding sensorimotor outcome, but regarding patient-reported outcome measures. J Plast Surg Hand Surg 2023; 57:95-102. [PMID: 34730072 DOI: 10.1080/2000656x.2021.1993868] [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: 10/19/2022]
Abstract
Despite the clinical importance of deep wrist injuries (DWIs), data comparing the outcome of suicide attempt survivors vs. accident survivors are lacking. Patients admitted to our Clinic for acute treatment of a DWI from 2008 to 2016 were contacted for a follow-up assessment of sensory, motor and functional outcomes. Patients also completed the Disability of the Arm, Shoulder and Hand Questionnaire, the Modified Mayo Wrist Score, the Boston Carpal Tunnel Questionnaire, and the WHOQOL-BREF questionnaires. 51 patients could be followed up, on average 4.3 ± 2.9 years after their injury. Suicide attempt survivors did not differ from accidents survivors concerning two-point discrimination, grip and pinch strength, but showed poorer outcomes in self-reported disability, symptom severity, and quality of life. Patients with DWIs from suicide attempts vs. accidents do not differ in sensorimotor outcomes but patient-reported outcome measures. Level of Evidence: II.
Collapse
Affiliation(s)
- Nico Matzkeit
- Clinic of Plastic Surgery, University of Lübeck, Lübeck, Germany
| | - Tobias Kisch
- Clinic of Plastic Surgery, University of Lübeck, Lübeck, Germany
| | - Annika Waldmann
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany.,Hamburg Cancer Registry, Ministry for Health and Consumer Protection, Hamburg, Germany
| | - Ulrich Schweiger
- Hamburg Cancer Registry, Ministry for Health and Consumer Protection, Hamburg, Germany
| | - Peter Mailänder
- Clinic of Plastic Surgery, University of Lübeck, Lübeck, Germany
| | - Anna Lisa Westermair
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Savari Kouzehkonan G, Motakef Kazemi N, Adabi M, Mosavi SE, Rezayat Sorkhabadi SM. Regeneration of sciatic nerve injury through nanofiber neural guidance channels containing collagen hydrogel and acetyl L carnitine: An in vitro and in vivo study. J BIOACT COMPAT POL 2022. [DOI: 10.1177/08839115221137654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Unlike the central nervous system (CNS), peripheral nervous system (PNS) injuries are partially repairable. Nerve guidance channels (NGCs) have been shown to improve the level of nerve repair after injury. In the present study, we developed a nanofiber NGC for the delivery of acetyl L carnitine (ALC) in a rat model of sciatic nerve injury. NGCs were produced by electrospinning a polymer blend of polycaprolacton and gelatin. The physicochemical and biological properties of developed scaffolds were investigated using Scanning electron microscopy, surface hydrophilicity measurement, porosity measurement, tensile strength studies, cell viability assay, and cell attachment assay. ALC was included in the collagen hydrogels at three weight ratios of 1%, 3%, and 5%. Cell viability assay showed that the hydrogels containing 5% ALC demonstrated a more favorable effect on PC-12 metabolic activity. Therefore, this concentration was chosen to treat PNS injury. The NGCs were implanted in rats and then their lumen was filled with collagen hydrogel + 5%ALC. The results of histopathological examinations and functional recovery studies showed that NGCs filled with ALC containing hydrogel have significant recovery potential compared to NGCs loaded with collagen hydrogels without ALC. Our results support the potential use of ALC-delivering NGCs in the treatment of peripheral nerve injury in the clinic.
Collapse
Affiliation(s)
- Gholamreza Savari Kouzehkonan
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negar Motakef Kazemi
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyedeh Elaheh Mosavi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Mahdi Rezayat Sorkhabadi
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
El Soury M, García-García ÓD, Tarulli I, Chato-Astrain J, Perroteau I, Geuna S, Raimondo S, Gambarotta G, Carriel V. Chitosan conduits enriched with fibrin-collagen hydrogel with or without adipose-derived mesenchymal stem cells for the repair of 15-mm-long sciatic nerve defect. Neural Regen Res 2022; 18:1378-1385. [PMID: 36453426 PMCID: PMC9838150 DOI: 10.4103/1673-5374.358605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Hollow conduits of natural or synthetic origins have shown acceptable regeneration results in short nerve gap repair; however, results are still not comparable with the current gold standard technique "autografts". Hollow conduits do not provide a successful regeneration outcome when it comes to critical nerve gap repair. Enriching the lumen of conduits with different extracellular materials and cells could provide a better biomimicry of the natural nerve regenerating environment and is expected to ameliorate the conduit performance. In this study, we evaluated nerve regeneration in vivo using hollow chitosan conduits or conduits enriched with fibrin-collagen hydrogels alone or with the further addition of adipose-derived mesenchymal stem cells in a 15 mm rat sciatic nerve transection model. Unexpected changes in the hydrogel consistency and structural stability in vivo led to a failure of nerve regeneration after 15 weeks. Nevertheless, the molecular assessment in the early regeneration phase (7, 14, and 28 days) has shown an upregulation of useful regenerative genes in hydrogel enriched conduits compared with the hollow ones. Hydrogels composed of fibrin-collagen were able to upregulate the expression of soluble NRG1, a growth factor that plays an important role in Schwann cell transdifferentiation. The further enrichment with adipose-derived mesenchymal stem cells has led to the upregulation of other important genes such as ErbB2, VEGF-A, BDNF, c-Jun, and ATF3.
Collapse
Affiliation(s)
- Marwa El Soury
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy,Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Óscar Darío García-García
- Department of Histology, Tissue Engineering Group, University of Granada, Granada, Spain,Instituto de Investigacion Biosanitaria, Ibs.GRANADA, Granada, Spain
| | - Isabella Tarulli
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Jesús Chato-Astrain
- Department of Histology, Tissue Engineering Group, University of Granada, Granada, Spain,Instituto de Investigacion Biosanitaria, Ibs.GRANADA, Granada, Spain
| | - Isabelle Perroteau
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy,Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy,Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy,Correspondence to: Stefania Raimondo, .
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy,Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Víctor Carriel
- Department of Histology, Tissue Engineering Group, University of Granada, Granada, Spain,Instituto de Investigacion Biosanitaria, Ibs.GRANADA, Granada, Spain
| |
Collapse
|
21
|
Trueman RP, Ahlawat AS, Phillips JB. A Shock to the (Nervous) System: Bioelectricity Within Peripheral Nerve Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1137-1150. [PMID: 34806913 DOI: 10.1089/ten.teb.2021.0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The peripheral nervous system has the remarkable ability to regenerate in response to injury. However, this is only successful over shorter nerve gaps and often provides poor outcomes for patients. Currently, the gold standard of treatment is the surgical intervention of an autograft, whereby patient tissue is harvested and transplanted to bridge the nerve gap. Despite being the gold standard, more than half of patients have dissatisfactory functional recovery after an autograft. Peripheral nerve tissue engineering aims to create biomaterials that can therapeutically surpass the autograft. Current tissue-engineered constructs are designed to deliver a combination of therapeutic benefits to the regenerating nerve, such as supportive cells, alignment, extracellular matrix, soluble factors, immunosuppressants, and other therapies. An emerging therapeutic opportunity in nerve tissue engineering is the use of electrical stimulation (ES) to modify and enhance cell function. ES has been shown to positively affect four key cell types, such as neurons, endothelial cells, macrophages, and Schwann cells, involved in peripheral nerve repair. Changes elicited include faster neurite extension, cellular alignment, and changes in cell phenotype associated with improved regeneration and functional recovery. This review considers the relevant modes of administration and cellular responses that could underpin incorporation of ES into nerve tissue engineering strategies. Impact Statement Tissue engineering is becoming increasingly complex, with multiple therapeutic modalities often included within the final tissue-engineered construct. Electrical stimulation (ES) is emerging as a viable therapeutic intervention to be included within peripheral nerve tissue engineering strategies; however, to date, there have been no review articles that collate the information regarding the effects of ES on key cell within peripheral nerve injury. This review article aims to inform the field on the different therapeutic effects that may be achieved by using ES and how they may become incorporated into existing strategies.
Collapse
Affiliation(s)
- Ryan P Trueman
- Center for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Ananya S Ahlawat
- Center for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - James B Phillips
- Center for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
22
|
Heinzel JC, Oberhauser V, Keibl C, Schädl B, Swiadek NV, Längle G, Frick H, Slezak C, Prahm C, Grillari J, Kolbenschlag J, Hercher D. ESWT Diminishes Axonal Regeneration following Repair of the Rat Median Nerve with Muscle-In-Vein Conduits but Not after Autologous Nerve Grafting. Biomedicines 2022; 10:biomedicines10081777. [PMID: 35892677 PMCID: PMC9394363 DOI: 10.3390/biomedicines10081777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 12/02/2022] Open
Abstract
Investigations reporting positive effects of extracorporeal shockwave therapy (ESWT) on nerve regeneration are limited to the rat sciatic nerve model. The effects of ESWT on muscle-in-vein conduits (MVCs) have also not been investigated yet. This study aimed to evaluate the effects of ESWT after repair of the rat median nerve with either autografts (ANGs) or MVCs. In male Lewis rats, a 7 mm segment of the right median nerve was reconstructed either with an ANG or an MVC. For each reconstructive technique, one group of animals received one application of ESWT while the other rats served as controls. The animals were observed for 12 weeks, and nerve regeneration was assessed using computerized gait analysis, the grasping test, electrophysiological evaluations and histological quantification of axons, blood vessels and lymphatic vasculature. Here, we provide for the first time a comprehensive analysis of ESWT effects on nerve regeneration in a rat model of median nerve injury. Furthermore, this study is among the first reporting the quantification of lymphatic vessels following peripheral nerve injury and reconstruction in vivo. While we found no significant direct positive effects of ESWT on peripheral nerve regeneration, results following nerve repair with MVCs were significantly inferior to those after ANG repair.
Collapse
Affiliation(s)
- Johannes C. Heinzel
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (J.C.H.); (C.P.); (J.K.)
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Viola Oberhauser
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Claudia Keibl
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Barbara Schädl
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Core Facility Morphology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Nicole V. Swiadek
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Gregor Längle
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Helen Frick
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Cyrill Slezak
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Physics, Utah Valley University, Orem, UT 84058, USA
| | - Cosima Prahm
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (J.C.H.); (C.P.); (J.K.)
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU—University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Jonas Kolbenschlag
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Klinik Tuebingen, University of Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (J.C.H.); (C.P.); (J.K.)
| | - David Hercher
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; (V.O.); (C.K.); (B.S.); (N.V.S.); (G.L.); (H.F.); (C.S.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Correspondence:
| |
Collapse
|
23
|
Hromada C, Hartmann J, Oesterreicher J, Stoiber A, Daerr A, Schädl B, Priglinger E, Teuschl-Woller AH, Holnthoner W, Heinzel J, Hercher D. Occurrence of Lymphangiogenesis in Peripheral Nerve Autografts Contrasts Schwann Cell-Induced Apoptosis of Lymphatic Endothelial Cells In Vitro. Biomolecules 2022; 12:820. [PMID: 35740945 PMCID: PMC9221261 DOI: 10.3390/biom12060820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
Peripheral nerve injuries pose a major clinical concern world-wide, and functional recovery after segmental peripheral nerve injury is often unsatisfactory, even in cases of autografting. Although it is well established that angiogenesis plays a pivotal role during nerve regeneration, the influence of lymphangiogenesis is strongly under-investigated. In this study, we analyzed the presence of lymphatic vasculature in healthy and regenerated murine peripheral nerves, revealing that nerve autografts contained increased numbers of lymphatic vessels after segmental damage. This led us to elucidate the interaction between lymphatic endothelial cells (LECs) and Schwann cells (SCs) in vitro. We show that SC and LEC secretomes did not influence the respective other cell types' migration and proliferation in 2D scratch assay experiments. Furthermore, we successfully created lymphatic microvascular structures in SC-embedded 3D fibrin hydrogels, in the presence of supporting cells; whereas SCs seemed to exert anti-lymphangiogenic effects when cultured with LECs alone. Here, we describe, for the first time, increased lymphangiogenesis after peripheral nerve injury and repair. Furthermore, our findings indicate a potential lymph-repellent property of SCs, thereby providing a possible explanation for the lack of lymphatic vessels in the healthy endoneurium. Our results highlight the importance of elucidating the molecular mechanisms of SC-LEC interaction.
Collapse
Affiliation(s)
- Carina Hromada
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; (C.H.); (A.D.); (A.H.T.-W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
| | - Jaana Hartmann
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Johannes Oesterreicher
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Anton Stoiber
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Anna Daerr
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; (C.H.); (A.D.); (A.H.T.-W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
| | - Barbara Schädl
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Eleni Priglinger
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Andreas H. Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria; (C.H.); (A.D.); (A.H.T.-W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
| | - Wolfgang Holnthoner
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| | - Johannes Heinzel
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Unfallklinik Tuebingen, University of Tuebingen, 72076 Tuebingen, Germany
| | - David Hercher
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (J.H.); (J.O.); (A.S.); (B.S.); (E.P.); (W.H.)
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria;
| |
Collapse
|
24
|
Adil A, Xu M, Haykal S. Recellularization of Bioengineered Scaffolds for Vascular Composite Allotransplantation. Front Surg 2022; 9:843677. [PMID: 35693318 PMCID: PMC9174637 DOI: 10.3389/fsurg.2022.843677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 05/09/2022] [Indexed: 12/20/2022] Open
Abstract
Traumatic injuries or cancer resection resulting in large volumetric soft tissue loss requires surgical reconstruction. Vascular composite allotransplantation (VCA) is an emerging reconstructive option that transfers multiple, complex tissues as a whole subunit from donor to recipient. Although promising, VCA is limited due to side effects of immunosuppression. Tissue-engineered scaffolds obtained by decellularization and recellularization hold great promise. Decellularization is a process that removes cellular materials while preserving the extracellular matrix architecture. Subsequent recellularization of these acellular scaffolds with recipient-specific cells can help circumvent adverse immune-mediated host responses and allow transplantation of allografts by reducing and possibly eliminating the need for immunosuppression. Recellularization of acellular tissue scaffolds is a technique that was first investigated and reported in whole organs. More recently, work has been performed to apply this technique to VCA. Additional work is needed to address barriers associated with tissue recellularization such as: cell type selection, cell distribution, and functionalization of the vasculature and musculature. These factors ultimately contribute to achieving tissue integration and viability following allotransplantation. The present work will review the current state-of-the-art in soft tissue scaffolds with specific emphasis on recellularization techniques. We will discuss biological and engineering process considerations, technical and scientific challenges, and the potential clinical impact of this technology to advance the field of VCA and reconstructive surgery.
Collapse
Affiliation(s)
- Aisha Adil
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Michael Xu
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of General Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Siba Haykal
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Plastic & Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Correspondence: Siba Haykal
| |
Collapse
|
25
|
Zhang G, Huang J, Hao S, Zhang J, Zhou N. Radix Astragalus Polysaccharide Accelerates Angiogenesis by Activating AKT/eNOS to Promote Nerve Regeneration and Functional Recovery. Front Pharmacol 2022; 13:838647. [PMID: 35431954 PMCID: PMC9010722 DOI: 10.3389/fphar.2022.838647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/04/2022] [Indexed: 11/17/2022] Open
Abstract
Peripheral nerve injury (PNI) results in loss of neural control and severe disabilities in patients. Promoting functional nerve recovery by accelerating angiogenesis is a promising neuroprotective treatment strategy. Here, we identified a bioactive Radix Astragalus polysaccharide (RAP) extracted from traditional Chinese medicine (TCM) as a potent enhancer of axonal regeneration and remyelination. Notably, RAP promoted functional recovery and delayed gastrocnemius muscle atrophy in a rat model of sciatic nerve crush injury. Further, RAP treatment may induce angiogenesis in vivo. Moreover, our in vitro results showed that RAP promotes endothelial cell (EC) migration and tube formation. Altogether, our results show that RAP can enhance functional recovery by accelerating angiogenesis, which was probably related to the activation of AKT/eNOS signaling pathway, thereby providing a polysaccharide-based therapeutic strategy for PNI.
Collapse
Affiliation(s)
- Geyi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinsheng Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuang Hao
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingchao Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Nan Zhou, ; Jingchao Zhang,
| | - Nan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Nan Zhou, ; Jingchao Zhang,
| |
Collapse
|
26
|
Angiogenesis is critical for the exercise-mediated enhancement of axon regeneration following peripheral nerve injury. Exp Neurol 2022; 353:114029. [DOI: 10.1016/j.expneurol.2022.114029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/17/2022] [Accepted: 02/27/2022] [Indexed: 11/21/2022]
|
27
|
Blood Vessels: The Pathway Used by Schwann Cells to Colonize Nerve Conduits. Int J Mol Sci 2022; 23:ijms23042254. [PMID: 35216370 PMCID: PMC8879195 DOI: 10.3390/ijms23042254] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 01/27/2023] Open
Abstract
The repair of severe nerve injuries requires an autograft or conduit to bridge the gap and avoid axon dispersion. Several conduits are used routinely, but their effectiveness is comparable to that of an autograft only for short gaps. Understanding nerve regeneration within short conduits could help improve their efficacy for longer gaps. Since Schwann cells are known to migrate on endothelial cells to colonize the “nerve bridge”, the new tissue spontaneously forming to connect the injured nerve stumps, here we aimed to investigate whether this migratory mechanism drives Schwann cells to also proceed within the nerve conduits used to repair large nerve gaps. Injured median nerves of adult female rats were repaired with 10 mm chitosan conduits and the regenerated nerves within conduits were analyzed at different time points using confocal imaging of sequential thick sections. Our data showed that the endothelial cells formed a dense capillary network used by Schwann cells to migrate from the two nerve stumps into the conduit. We concluded that angiogenesis played a key role in the nerve conduits, not only by supporting cell survival but also by providing a pathway for the migration of newly formed Schwann cells.
Collapse
|
28
|
Heparin Immobilization of Tissue Engineered Xenogeneic Small Diameter Arterial Scaffold Improve Endothelialization. Tissue Eng Regen Med 2022; 19:505-523. [PMID: 35092597 PMCID: PMC9130405 DOI: 10.1007/s13770-021-00411-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Autologous vessels graft (Inner diameter < 6 mm) harvesting always challenged during bypass grafting surgery and its complication shows poor outcome. Tissue engineered vascular graft allow to generate biological graft without any immunogenic complication. The approach presented in this study is to induce graft remodeling through heparin coating in luminal surface of small diameter (Inner diameter < 1 mm) decellularized arterial graft. METHODS Decellularization of graft was done using SDS, combination of 0.5% sodium dodecyl sulfate and 0.5% sodium deoxycholate and only sodium deoxycholate. Decellularization was confirmed on basis of histology, and DAPI. Characterization of extracellular matrix was analyzed using histology and scanning electron microscopy. Surface modification of decellularized vascular graft was done with heparin coating. Heparin immobilization was evaluated by toluidine blue stain. Heparin-coated graft was transplanted end to end anastomosis in femoral artery in rat. RESULTS Combination of 0.5% sodium dodecyl sulfate and 0.5% Sodium deoxycholate showed complete removal of xenogeneic cells. The heparin coating on luminal surface showed anti-thrombogenicity and endothelialization. Mechanical testing revealed no significant differences in strain characteristics and modulus between native tissues, decellularized scaffolds and transplanted scaffold. Collectively, this study proposed a heparin-immobilized ECM coating to surface modification offering functionalize biomaterials for developing small-diameter vascular grafts. CONCLUSION We conclude that xenogeneic decellularized arterial scaffold with heparin surface modification can be fabricated and successfully transplanted small diameter (inner diameter < 1 mm) decellularized arterial graft.
Collapse
|
29
|
Takeuchi H, Ikeguchi R, Noguchi T, Ando M, Yoshimoto K, Sakamoto D, Matsuda S. The efficacy of combining a vascularized biogenic conduit and a decellularized nerve graft in the treatment of peripheral nerve defects: An experimental study using the rat sciatic nerve defect model. Microsurgery 2021; 42:254-264. [PMID: 34953149 DOI: 10.1002/micr.30853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/09/2021] [Accepted: 11/29/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Although decellularized nerve grafts are often used as a bridging material in nerve defect repair, the lack of perfusion support in this procedure limits the regeneration capacity. To address this, we applied vascularized biogenic conduits, which are fibrous membranes prefabricated around the silicone rod that contain rich vascularity and macrophages, to nerve defect repair procedures using decellularized nerve grafts. The purpose of this study is to investigate the capacity of combining a vascularized biogenic conduit and a decellularized nerve graft for peripheral nerve regeneration using a 10-mm nerve defect model in rats. MATERIALS AND METHODS Sixteen adult male rats (F344 rats, 10-12 weeks, 200-250 g) were used in this study. For the prefabrication of vascularized biogenic conduits, a silicone rod was transplanted next to the sciatic nerve. After 8 weeks, this silicone rod was enveloped in connective tissue, called a vascularized biogenic conduit. The first rat was used to investigate the histological characteristics of vascularized biogenic conduits through immunofluorescence studies. The remaining 15 rats were divided into three groups to evaluate the efficacy of the combination of a decellularized nerve graft and a vascularized biogenic conduit: a decellularized nerve graft (DNG) group, a decellularized nerve graft with a vascularized biogenic conduit (DNG-w-VBC) group, and an autologous nerve graft (ANG) group. Eight weeks after nerve graft surgery, the assessment results of both functional recovery (electrophysical studies and target muscle atrophy) and morphological recovery (total number, diameter, and myelin thickness of the regenerated axons) of the regenerated nerves were examined. RESULTS Immunofluorescence studies revealed that the VBC contains extracellular matrix, vascular tissue, and macrophages. The results of the DNG-w-VBC group were superior to the DNG group in electrophysiological studies (CMAP; 6.29 ± 0.80% vs. 4.02 ± 3.35%, MNCV; 50.6 ± 8.4% vs. 25.7 ± 15.6%, p < .05, respectively), regenerated axon number (11,348 ± 812 vs. 7697 ± 2197, p < .05), and mean axon diameter (2.72 ± 0.33 μm vs. 1.64 ± 0.12 μm, p < .05). CONCLUSIONS Our study confirms that vascularized biogenic conduits supply vascularity and macrophages to nerve defect sites. Combining vascularized biogenic conduits with decellularized nerve grafts to treat nerve defects offers superior functional and morphological recovery of regenerated axons.
Collapse
Affiliation(s)
- Hisataka Takeuchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Maki Ando
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Yoshimoto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daichi Sakamoto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
30
|
Repair of peripheral nerve injuries using a prevascularized cell-based tissue-engineered nerve conduit. Biomaterials 2021; 280:121269. [PMID: 34847434 DOI: 10.1016/j.biomaterials.2021.121269] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/09/2021] [Accepted: 11/21/2021] [Indexed: 12/15/2022]
Abstract
One of the major challenges in the development of a larger and longer nerve conduit for peripheral nerve repair is the limitation in oxygen and nutrient diffusion within the tissue after transplantation preventing Schwann cell and axonal migration. This restriction is due to the slow neovascularization process of the graft starting from both nerve endings. To overcome this limitation, we propose the design of a living tissue-engineered nerve conduit made of an internal tube with a three-dimensional structure supporting axonal migration, which is inserted inside a hollow external tube that plays the role of an epineurium and is strong enough to be stitched to the severed nerve stumps. The internal tube is made of a rolled living fibroblast sheet and can be seeded with endothelial cells to promote the formation of a network containing capillary-like structures which allow rapid inosculation with the host nerve microvasculature after grafting. Human nerve conduits were grafted in immunodeficient rats to bridge a 15 mm sciatic nerve gap. Human capillaries within the pre-vascularized nerve conduit successfully connected to the host circulation 2 weeks after grafting. Twenty-two weeks after surgery, rats transplanted with the nerve conduits had a similar motor function recovery compared to the autograft group. By promoting rapid vascularization of the internal nerve tube from both ends of the nerve stumps, this endothelialized nerve conduit model displays a favorable environment to enhance axonal migration in both larger caliber and longer nerve grafts.
Collapse
|
31
|
Shen J, Wang J, Liu X, Sun Y, Yin A, Chai Y, Zhang K, Wang C, Zheng X. In Situ Prevascularization Strategy with Three-Dimensional Porous Conduits for Neural Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50785-50801. [PMID: 34664947 DOI: 10.1021/acsami.1c16138] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Neovascularization is crucial for peripheral nerve regeneration and long-term functional restoration. Previous studies have emphasized strategies that enhance axonal repair over vascularization. Here, we describe the development and application of an in situ prevascularization strategy that uses 3D porous nerve guidance conduits (NGCs) to achieve angiogenesis-mediated neural regeneration. The optimal porosity of the NGC is a critical feature for achieving neovascularization and nerve growth patency. Hollow silk fibroin/poly(l-lactic acid-co-ε-caprolactone) NGCs with 3D sponge-like walls were fabricated using electrospinning and freeze-drying. In vitro results showed that 3D porous NGC favored cell biocompatibility had neuroregeneration potential and, most importantly, had angiogenic activity. Results from our mechanistic studies suggest that activation of HIF-1α signaling might be associated with this process. We also tested in situ prevascularized 3D porous NGCs in vivo by transplanting them into a 10 mm rat sciatic nerve defect model with the aim of regenerating the severed nerve. The prevascularized 3D porous NGCs greatly enhanced intraneural angiogenesis, resulting in demonstrable neurogenesis. Eight weeks after transplantation, the performance of the prevascularized 3D NGCs was similar to that of traditional autografts in terms of improved anatomical structure, morphology, and neural function. In conclusion, combining a reasonably fabricated 3D-pore conduit structure with in situ prevascularization promoted functional nerve regeneration, suggesting an alternative strategy for achieving functional recovery after peripheral nerve trauma.
Collapse
Affiliation(s)
- Junjie Shen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
- Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Hainan 570300, PR China
| | - Jiayan Wang
- College of Materials and Textile Engineering, Nanotechnology Research Institute, Jiaxing University, Zhejiang 314001, PR China
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology, Zhejiang 314001, PR China
| | - Xuanzhe Liu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| | - Yi Sun
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| | - Anlin Yin
- College of Materials and Textile Engineering, Nanotechnology Research Institute, Jiaxing University, Zhejiang 314001, PR China
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology, Zhejiang 314001, PR China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| | - Kuihua Zhang
- College of Materials and Textile Engineering, Nanotechnology Research Institute, Jiaxing University, Zhejiang 314001, PR China
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology, Zhejiang 314001, PR China
| | - Chunyang Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
- Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Hainan 570300, PR China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| |
Collapse
|
32
|
Chen S, Zhu J, Zhang Y, Cai X, Yi S, Wang X. miR-328a-3p stimulates endothelial cell migration and tubulogenesis. Exp Ther Med 2021; 22:1104. [PMID: 34504558 PMCID: PMC8383776 DOI: 10.3892/etm.2021.10538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022] Open
Abstract
Endothelial cells have important biological roles after peripheral nerve injury by forming blood vessels within the nerve gap and guiding Schwann cell migration. MicroRNAs (miRNAs/miRs) affect cellular behavior and regulate a wide variety of physiological and pathological activities, including peripheral nerve regeneration. Emerging studies have identified the essential roles of miRNAs in the phenotype modulation of Schwann cells, while the effects of miRNAs on endothelial cells have remained to be thoroughly investigated. miR-328a-3p was differentially expressed in peripheral nerve stumps after nerve injury. In the present study, the effects of miR-328a-3p on biological functions of endothelial cells were determined by transfecting cultured human umbilical vein endothelial cells (HUVECs) with miR-328a-3p mimics or inhibitor. Transfection with miR-328a-3p mimics led to slightly decreased HUVEC proliferation and robustly increased HUVEC migration and tubulogenesis, while transfection with miR-328a-3p inhibitor led to opposite results. Using bioinformatics analysis, potential regulators and effectors of miR-328a-3p were further discovered and a miR-328a-3p-centered competing endogenous RNA network was constructed. Collectively, the present study demonstrated that dysregulated miR-328a-3p after peripheral nerve injury may affect the migration and angiogenesis of endothelial cells and contribute to peripheral nerve regeneration.
Collapse
Affiliation(s)
- Sailing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jun Zhu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yunsong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaodong Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
33
|
Malheiro A, Wieringa P, Moroni L. Peripheral neurovascular link: an overview of interactions and in vitro models. Trends Endocrinol Metab 2021; 32:623-638. [PMID: 34127366 DOI: 10.1016/j.tem.2021.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Nerves and blood vessels (BVs) establish extensive arborized networks to innervate tissues and deliver oxygen/metabolic support. Developmental cues direct the formation of these intricate and often overlapping patterns, which reflect close interactions within the peripheral neurovascular system. Besides the mutual dependence to survive and function, nerves and BVs share several receptors and ligands, as well as principles of differentiation, growth and pathfinding. Neurovascular (NV) interactions are maintained in adult life and are essential for certain regenerative mechanisms, such as wound healing. In pathological situations (e.g., type 2 diabetes mellitus), the NV system can be severely perturbed and become dysfunctional. Unwanted neural growth and vascularization are also associated with the progression of some pathologies, such as cancer and endometriosis. In this review, we describe the fundamental NV interactions in development, highlighting the similarities between both networks and wiring mechanisms. We also describe the NV contribution to regenerative processes and potential pathological dysfunctions. Finally, we provide an overview of current in vitro models used to replicate and investigate the NV ecosystem, addressing present limitations and future perspectives.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands.
| |
Collapse
|
34
|
Surgical Angiogenesis of Decellularized Nerve Allografts Improves Early Functional Recovery in a Rat Sciatic Nerve Defect Model. Plast Reconstr Surg 2021; 148:561-570. [PMID: 34292916 DOI: 10.1097/prs.0000000000008291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Surgical angiogenesis applied to nerve grafts has been suggested to enhance nerve regeneration after nerve injury. The authors hypothesized that surgical angiogenesis to decellularized nerve allografts would improve functional recovery in a rat sciatic nerve defect model. METHODS Sixty Lewis rats were divided in three groups of 20 animals each. Unilateral sciatic nerve defects were repaired with (1) autografts, (2) decellularized allografts, and (3) decellularized allografts wrapped with a superficial inferior epigastric artery fascial flap to add surgical angiogenesis. Twelve and 16 weeks after surgery, nerve regeneration was assessed using functional, electrophysiologic, histologic, and immunofluorescence analyses. Ultrasonography was used during the survival period to noninvasively evaluate muscle atrophy and reinnervation by measuring cross-sectional muscle area. RESULTS Surgical angiogenesis of allografts demonstrated significantly improved isometric tetanic force recovery at 12 weeks, compared to allograft alone, which normalized between groups at 16 weeks. Cross-sectional muscle areas showed no differences between groups. Electrophysiology showed superiority of autografts at both time points. No differences were found in histologic analysis, besides a significantly inferior N ratio in allografts at 12 weeks. Immunofluorescent expression of CD34, indicating vascularity, was significantly enhanced in the superficial inferior epigastric artery fascial group compared to allografts at 12 weeks, with highest expression at 16 weeks compared to all groups. CONCLUSION Surgical angiogenesis with an adipofascial flap to the nerve allograft increases vascularity in the nerve graft, with subsequent improvement of early muscle force recovery, comparable to autografts.
Collapse
|
35
|
Wang J, Xu X, Dong W. Correlation Analysis Between Local Cerebral Blood Flow and Severity of Vascular Cognitive Dysfunction. JOURNAL OF MEDICAL IMAGING AND HEALTH INFORMATICS 2021. [DOI: 10.1166/jmihi.2021.3529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Research methods: This paper analyses the correlation between cerebral blood flow perfusion caused by cerebral vascular stenosis and the reduction of patients with cognitive dysfunction and white matter damage. A total of 118 patients with reduced cerebral blood flow perfusion
due to cerebrovascular stenosis were selected to be included in the disease group, and 118 patients with no cerebrovascular stenosis and no neurological disease were included in the control group. The cerebral blood flow perfusion index and cognitive function index were compared between the
two groups of patients. The correlation between each index and the degree of brain white matter damage was analysed. Results: The scores of brain white matter damage in patients with disease group were higher than those in control group, and cCBV, cCBF, TTP, MTT, MoCA, MMSE, ADL, and
WMS were lower than those in control group, and the difference was statistically significant (P < 0.05). cCBV, cCBF, TTP, MTT, and white matter damage scores were highly correlated with MoCA, MMSE, ADL, and WMS (P < 0.05). There is a clear correlation between cerebral vascular
perfusion, cognitive dysfunction, and white matter damage in patients with cerebrovascular stenosis. The more severe the perfusion of cerebral blood flow, the more severe the cognitive dysfunction and the white matter damage.
Collapse
Affiliation(s)
- Jinling Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xin Xu
- Department of Neurology, Daqing Oilfield General Hospital, Zhongkang Street, Daqing, Heilongjiang, 163001, China
| | - Wanhui Dong
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| |
Collapse
|
36
|
Comprehensive strategy of conduit guidance combined with VEGF producing Schwann cells accelerates peripheral nerve repair. Bioact Mater 2021; 6:3515-3527. [PMID: 33842738 PMCID: PMC8008177 DOI: 10.1016/j.bioactmat.2021.03.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/18/2021] [Accepted: 03/06/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve regeneration requires stepwise and well-organized establishment of microenvironment. Since local delivery of VEGF-A in peripheral nerve repair is expected to promote angiogenesis in the microenvironment and Schwann cells (SCs) play critical role in nerve repair, combination of VEGF and Schwann cells may lead to efficient peripheral nerve regeneration. VEGF-A overexpressing Schwann cells were established and loaded into the inner wall of hydroxyethyl cellulose/soy protein isolate/polyaniline sponge (HSPS) conduits. When HSPS is mechanically distorted, it still has high durability of strain strength, thus, can accommodate unexpected strain of nerve tissues in motion. A 10 mm nerve defect rat model was used to test the repair performance of the HSPS-SC (VEGF) conduits, meanwhile the HSPS, HSPS-SC, HSPS-VEGF conduits and autografts were worked as controls. The immunofluorescent co-staining of GFP/VEGF-A, Ki67 and MBP showed that the VEGF-A overexpressing Schwann cells could promote the proliferation, migration and differentiation of Schwann cells as the VEGF-A was secreted from the VEGF-A overexpressing Schwann cells. The nerve repair performance of the multifunctional and flexible conduits was examined though rat behavioristics, electrophysiology, nerve innervation to gastrocnemius muscle (GM), toluidine blue (TB) staining, transmission electron microscopy (TEM) and NF200/S100 double staining in the regenerated nerve. The results displayed that the effects on the repair of peripheral nerves in HSPS-SC (VEGF) group was the best among the conduits groups and closed to autografts. HSPS-SC (VEGF) group exhibited notably increased CD31+ endothelial cells and activation of VEGFR2/ERK signaling pathway in the regenerated nerve tissues, which probably contributed to the improved nerve regeneration. Altogether, the comprehensive strategy including VEGF overexpressing Schwann cells-mediated and HSPS conduit-guided peripheral nerve repair provides a new avenue for nerve tissue engineering.
Collapse
|
37
|
Dömer P, Kayal J, Janssen-Bienhold U, Kewitz B, Kretschmer T, Heinen C. Rapid and efficient immunomagnetic isolation of endothelial cells from human peripheral nerves. Sci Rep 2021; 11:1951. [PMID: 33479384 PMCID: PMC7820485 DOI: 10.1038/s41598-021-81361-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/04/2021] [Indexed: 01/18/2023] Open
Abstract
Endothelial cells (ECs) have gained an increased scientific focus since they were reported to provide guidance for Schwann cells and subsequently following axons after nerve injuries. However, previous protocols for the isolation of nerve-derived ECs from human nerves are ineffective regarding time and yield. Therefore, we established a novel and efficient protocol for the isolation of ECs from human peripheral nerves by means of immunomagnetic CD31-antibody conjugated Dynabeads and assessed the purity of the isolated cells. The easy-to-follow and time-effective isolation method allows the isolation of > 95% pure ECs. The isolated ECs were shown to express highly specific EC marker proteins and revealed functional properties by formation of CD31 and VE-cadherin positive adherens junctions, as well as ZO-1 positive tight-junctions. Moreover, the formation of capillary EC-tubes was observed in-vitro. The novel protocol for the isolation of human nerve-derived ECs allows and simplifies the usage of ECs in research of the human blood-nerve-barrier and peripheral nerve regeneration. Additionally, a potential experimental application of patient-derived nerve ECs in the in-vitro vascularization of artificial nerve grafts is feasible.
Collapse
Affiliation(s)
- Patrick Dömer
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany.
- Department of Neurosurgery, Evangelisches Krankenhaus, Campus Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| | - Janine Kayal
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany
| | - Ulrike Janssen-Bienhold
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Bettina Kewitz
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany
- Department of Neurosurgery, Evangelisches Krankenhaus, Campus Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Thomas Kretschmer
- Department of Neurosurgery and Neurorestauration, Klinikum Klagenfurt Am Wörthersee, Klagenfurt, Austria
| | - Christian Heinen
- Department of Neurosurgery, Evangelisches Krankenhaus, Campus Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
38
|
Fundamentals and Current Strategies for Peripheral Nerve Repair and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1249:173-201. [PMID: 32602098 DOI: 10.1007/978-981-15-3258-0_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A body of evidence indicates that peripheral nerves have an extraordinary yet limited capacity to regenerate after an injury. Peripheral nerve injuries have confounded professionals in this field, from neuroscientists to neurologists, plastic surgeons, and the scientific community. Despite all the efforts, full functional recovery is still seldom. The inadequate results attained with the "gold standard" autograft procedure still encourage a dynamic and energetic research around the world for establishing good performing tissue-engineered alternative grafts. Resourcing to nerve guidance conduits, a variety of methods have been experimentally used to bridge peripheral nerve gaps of limited size, up to 30-40 mm in length, in humans. Herein, we aim to summarize the fundamentals related to peripheral nerve anatomy and overview the challenges and scientific evidences related to peripheral nerve injury and repair mechanisms. The most relevant reports dealing with the use of both synthetic and natural-based biomaterials used in tissue engineering strategies when treatment of nerve injuries is envisioned are also discussed in depth, along with the state-of-the-art approaches in this field.
Collapse
|
39
|
Adipose-Derived Stem Cells: Current Applications and Future Directions in the Regeneration of Multiple Tissues. Stem Cells Int 2020; 2020:8810813. [PMID: 33488736 PMCID: PMC7787857 DOI: 10.1155/2020/8810813] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/04/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) can maintain self-renewal and enhanced multidifferentiation potential through the release of a variety of paracrine factors and extracellular vesicles, allowing them to repair damaged organs and tissues. Consequently, considerable attention has increasingly been paid to their application in tissue engineering and organ regeneration. Here, we provide a comprehensive overview of the current status of ADSC preparation, including harvesting, isolation, and identification. The advances in preclinical and clinical evidence-based ADSC therapy for bone, cartilage, myocardium, liver, and nervous system regeneration as well as skin wound healing are also summarized. Notably, the perspectives, potential challenges, and future directions for ADSC-related researches are discussed. We hope that this review can provide comprehensive and standardized guidelines for the safe and effective application of ADSCs to achieve predictable and desired therapeutic effects.
Collapse
|
40
|
Nanoscience and nanotechnology in fabrication of scaffolds for tissue regeneration. INTERNATIONAL NANO LETTERS 2020. [DOI: 10.1007/s40089-020-00318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
41
|
Qi Y, Zou T, Dissanayaka WL, Wong HM, Bertassoni LE, Zhang C. Fabrication of Tapered Fluidic Microchannels Conducive to Angiogenic Sprouting within Gelatin Methacryloyl Hydrogels. J Endod 2020; 47:52-61. [PMID: 33045266 DOI: 10.1016/j.joen.2020.08.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The transplantation of stem cells/tissue constructs into root canal space is a promising strategy for regenerating lost pulp tissue. However, the root canal system, which is cone shaped with a taper from the larger coronal end to the smaller apical end, limits the vascular supply and, therefore, the regenerative capacity. The current study aimed to fabricate built-in microchannels with different tapers to explore various approaches to endothelialize these microchannels. METHODS The fluidic microchannels with varying tapers (parallel, 0.04, and 0.06) were fabricated within gelatin methacryloyl (GelMA) hydrogel (with or without stem cell from the apical papilla [SCAP] encapsulation) of different concentrations (5%, 7.5%, and 10% [w/v]). Green fluorescent protein-expressing human umbilical vein endothelial cells (HUVECs-GFP) were seeded alone or with SCAPs in coculture into these microchannels. Angiogenic sprouting was assessed by fluorescence and a confocal microscope and ImageJ software (National Institutes of Health, Bethesda, MD). Immunostaining was conducted to illustrate monolayer formation. Data were statistically analyzed by 1-way/2-way analysis of variance. RESULTS HUVEC-only inoculation formed an endothelial monolayer inside the microchannel without angiogenic sprouting. HUVECs-GFP/SCAPs cocultured at a 1:1 ratio produced the longest sprouting compared with the other 3 ratios. The average length of the sprouting in the 0.04 taper microchannel was significantly longer compared with that in the parallel and 0.06 taper microchannels. Significant differences in HUVEC-GFP sprouting were observed in 5% GelMA hydrogel. Encapsulation of SCAPs within hydrogel further stimulated the sprouting of HUVECs. CONCLUSIONS The coculture of SCAPs and HUVECs-GFP at a ratio of 1:1 in 0.04 taper fluidic microchannels fabricated with 5% (w/v) GelMA hydrogel with SCAPs encapsulated was found to be the optimal condition to enhance angiogenesis inside tapered microchannels.
Collapse
Affiliation(s)
- Yubingqing Qi
- Department of Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ting Zou
- Department of Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Waruna Lakmal Dissanayaka
- Department of Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hai Ming Wong
- Department of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Luiz E Bertassoni
- Department of Biomaterials and Biomechanics, School of Dentistry Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Chengfei Zhang
- Department of Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
42
|
Mehrabi T, Mesgar AS, Mohammadi Z. Bioactive Glasses: A Promising Therapeutic Ion Release Strategy for Enhancing Wound Healing. ACS Biomater Sci Eng 2020; 6:5399-5430. [PMID: 33320556 DOI: 10.1021/acsbiomaterials.0c00528] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The morbidity, mortality, and burden of burn victims and patients with severe diabetic wounds are still high, which leads to an extensively growing demand for novel treatments with high clinical efficacy. Biomaterial-based wound treatment approaches have progressed over time from simple cotton wool dressings to advanced skin substitutes containing cells and growth factors; however, no wound care approach is yet completely satisfying. Bioactive glasses are materials with potential in many areas that exhibit unique features in biomedical applications. Today, bioactive glasses are not only amorphous solid structures that can be used as a substitute in hard tissue but also are promising materials for soft tissue regeneration and wound healing applications. Biologically active elements such as Ag, B, Ca, Ce, Co, Cu, Ga, Mg, Se, Sr, and Zn can be incorporated in glass networks; hence, the superiority of these multifunctional materials over current materials results from their ability to release multiple therapeutic ions in the wound environment, which target different stages of the wound healing process. Bioactive glasses and their dissolution products have high potency for inducing angiogenesis and exerting several biological impacts on cell functions, which are involved in wound healing and some other features that are valuable in wound healing applications, namely hemostatic and antibacterial properties. In this review, we focus on skin structure, the dynamic process of wound healing in injured skin, and existing wound care approaches. The basic concepts of bioactive glasses are reviewed to better understand the relationship between glass structure and its properties. We illustrate the active role of bioactive glasses in wound repair and regeneration. Finally, research studies that have used bioactive glasses in wound healing applications are summarized and the future trends in this field are elaborated.
Collapse
Affiliation(s)
- Tina Mehrabi
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Abdorreza S Mesgar
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Zahra Mohammadi
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| |
Collapse
|
43
|
Muangsanit P, Day A, Dimiou S, Ataç AF, Kayal C, Park H, Nazhat SN, Phillips JB. Rapidly formed stable and aligned dense collagen gels seeded with Schwann cells support peripheral nerve regeneration. J Neural Eng 2020; 17:046036. [DOI: 10.1088/1741-2552/abaa9c] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
Development of vascularized nerve scaffold using perfusion-decellularization and recellularization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111311. [PMID: 32919672 DOI: 10.1016/j.msec.2020.111311] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 07/15/2020] [Accepted: 07/25/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Vascularized nerve grafts (VNG) may offer an advantage in peripheral nerve regeneration by avoiding ischemic damage and central necrosis observed in non-VNG, particularly for the treatment of large and long nerve defects. However, surgical complexity, donor site morbidity and limited nerve availability remain important drawbacks for the clinical use of VNG. Here we explore the potential of perfusion-decellularization for bioengineering a VNG to be used in peripheral nerve reconstruction. METHODS Porcine sciatic nerves were surgically procured along with their vascular pedicle attached. The specimens were decellularized via perfusion-decellularization and preservation of the extracellular matrix (ECM), vascular patency and tissue cytokine contents were examined. Scaffold reendothelialization was conducted with porcine aortic endothelial cells in a perfusion-bioreactor. RESULTS Morphologic examination of decellularized VNG and analysis of the DNA content demonstrated cell clearance whereas ECM content and structures of the nerve fascicles were preserved. Using 3D micro-computed tomography imaging we observed optimal vasculature preservation in decellularized scaffolds, down to the capillary level. Cytokine quantification demonstrated measurable levels of growth factors after decellularization. Endothelial cell engraftment of the large caliber vessels was observed in reendothelialized scaffolds. CONCLUSIONS In this study we provide evidence that perfusion-decellularization can be used to create vascularized nerve scaffolds in which the vasculature and the ECM component are well preserved. As compared to non-vascularized conduits, engineered vascularized nerve scaffolds may represent an ideal approach for promoting better nerve regeneration in larger nerve defect reconstructions.
Collapse
|
45
|
Hao D, Swindell HS, Ramasubramanian L, Liu R, Lam KS, Farmer DL, Wang A. Extracellular Matrix Mimicking Nanofibrous Scaffolds Modified With Mesenchymal Stem Cell-Derived Extracellular Vesicles for Improved Vascularization. Front Bioeng Biotechnol 2020; 8:633. [PMID: 32671037 PMCID: PMC7329993 DOI: 10.3389/fbioe.2020.00633] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
The network structure and biological components of natural extracellular matrix (ECM) are indispensable for promoting tissue regeneration. Electrospun nanofibrous scaffolds have been widely used in regenerative medicine to provide structural support for cell growth and tissue regeneration due to their natural ECM mimicking architecture, however, they lack biological functions. Extracellular vesicles (EVs) are potent vehicles of intercellular communication due to their ability to transfer RNAs, proteins, and lipids, thereby mediating significant biological functions in different biological systems. Matrix-bound nanovesicles (MBVs) are identified as an integral and functional component of ECM bioscaffolds mediating significant regenerative functions. Therefore, to engineer EVs modified electrospun scaffolds, mimicking the structure of the natural EV-ECM complex and the physiological interactions between the ECM and EVs, will be attractive and promising in tissue regeneration. Previously, using one-bead one-compound (OBOC) combinatorial technology, we identified LLP2A, an integrin α4β1 ligand, which had a strong binding to human placenta-derived mesenchymal stem cells (PMSCs). In this study, we isolated PMSCs derived EVs (PMSC-EVs) and demonstrated they expressed integrin α4β1 and could improve endothelial cell (EC) migration and vascular sprouting in an ex vivo rat aortic ring assay. LLP2A treated culture surface significantly improved PMSC-EV attachment, and the PMSC-EV treated culture surface significantly enhanced the expression of angiogenic genes and suppressed apoptotic activity. We then developed an approach to enable "Click chemistry" to immobilize LLP2A onto the surface of electrospun scaffolds as a linker to immobilize PMSC-EVs onto the scaffold. The PMSC-EV modified electrospun scaffolds significantly promoted EC survival and angiogenic gene expression, such as KDR and TIE2, and suppressed the expression of apoptotic markers, such as caspase 9 and caspase 3. Thus, PMSC-EVs hold promising potential to functionalize biomaterial constructs and improve the vascularization and regenerative potential. The EVs modified biomaterial scaffolds can be widely used for different tissue engineering applications.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Hila Shimshi Swindell
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Lalithasri Ramasubramanian
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
46
|
Fibroblasts Colonizing Nerve Conduits Express High Levels of Soluble Neuregulin1, a Factor Promoting Schwann Cell Dedifferentiation. Cells 2020; 9:cells9061366. [PMID: 32492853 PMCID: PMC7349576 DOI: 10.3390/cells9061366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/26/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
Conduits for the repair of peripheral nerve gaps are a good alternative to autografts as they provide a protected environment and a physical guide for axonal re-growth. Conduits require colonization by cells involved in nerve regeneration (Schwann cells, fibroblasts, endothelial cells, macrophages) while in the autograft many cells are resident and just need to be activated. Since it is known that soluble Neuregulin1 (sNRG1) is released after injury and plays an important role activating Schwann cell dedifferentiation, its expression level was investigated in early regeneration steps (7, 14, 28 days) inside a 10 mm chitosan conduit used to repair median nerve gaps in Wistar rats. In vivo data show that sNRG1, mainly the isoform α, is highly expressed in the conduit, together with a fibroblast marker, while Schwann cell markers, including NRG1 receptors, were not. Primary culture analysis shows that nerve fibroblasts, unlike Schwann cells, express high NRG1α levels, while both express NRG1β. These data suggest that sNRG1 might be mainly expressed by fibroblasts colonizing nerve conduit before Schwann cells. Immunohistochemistry analysis confirmed NRG1 and fibroblast marker co-localization. These results suggest that fibroblasts, releasing sNRG1, might promote Schwann cell dedifferentiation to a “repair” phenotype, contributing to peripheral nerve regeneration.
Collapse
|
47
|
Bin Z, Zhihu Z, Jianxiong M, Xinlong M. Repairing peripheral nerve defects with revascularized tissue-engineered nerve based on a vascular endothelial growth factor-heparin sustained release system. J Tissue Eng Regen Med 2020; 14:819-828. [PMID: 32336028 DOI: 10.1002/term.3048] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/30/2020] [Accepted: 04/11/2020] [Indexed: 12/30/2022]
Abstract
To enhance the angiogenic capacity of tissue-engineered peripheral nerves, we have constructed revascularized tissue-engineered nerves based on a vascular endothelial growth factor (VEGF)-heparin sustained release system. However, the effects of the repair of large peripheral nerve defects are not known. In this study, we used the above revascularized tissue-engineered nerve to repair large nerve defects in rats. The repair effects were observed through general observation, functional evaluation of nerve regeneration, ultrasound examination, neural electrophysiology, wet weight ratio of bilateral gastrocnemius muscle, histological evaluation, and quantitative real-time polymerase chain reaction (PCR) analysis. The results showed that the tissue-engineered peripheral nerve based on a VEGF-heparin sustained release system can achieve early vascularization and restore blood supply in the nerve graft area. The realization of early vascularization in the area of the nerve defect greatly promotes the speed of nerve regeneration and reconstruction in the area of the nerve defect, which greatly advances the process of nerve repair and reconstruction and accelerates the restoration of the normal morphological structure and function of peripheral nerves.
Collapse
Affiliation(s)
- Zhao Bin
- Institute of Orthopedics, Tianjin Hospital, Tianjin, China
| | - Zhao Zhihu
- Institute of Orthopedics, Tianjin Hospital, Tianjin, China
| | - Ma Jianxiong
- Institute of Orthopedics, Tianjin Hospital, Tianjin, China
| | - Ma Xinlong
- Institute of Orthopedics, Tianjin Hospital, Tianjin, China
| |
Collapse
|
48
|
Gaihre B, Liu X, Lee Miller A, Yaszemski M, Lu L. Poly(Caprolactone Fumarate) and Oligo[Poly(Ethylene Glycol) Fumarate]: Two Decades of Exploration in Biomedical Applications. POLYM REV 2020. [DOI: 10.1080/15583724.2020.1758718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - A. Lee Miller
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Yaszemski
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
49
|
Saffari TM, Badreldin A, Mathot F, Bagheri L, Bishop AT, van Wijnen AJ, Shin AY. Surgical angiogenesis modifies the cellular environment of nerve allografts in a rat sciatic nerve defect model. Gene 2020; 751:144711. [PMID: 32353583 DOI: 10.1016/j.gene.2020.144711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/13/2020] [Accepted: 04/23/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Tiam M Saffari
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Plastic, Reconstructive and Hand Surgery, Radboud University, Nijmegen, The Netherlands
| | - Amr Badreldin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Femke Mathot
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Plastic, Reconstructive and Hand Surgery, Radboud University, Nijmegen, The Netherlands
| | - Leila Bagheri
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Allen T Bishop
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| | - Alexander Y Shin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
50
|
Fang Z, Ge X, Chen X, Xu Y, Yuan WE, Ouyang Y. Enhancement of sciatic nerve regeneration with dual delivery of vascular endothelial growth factor and nerve growth factor genes. J Nanobiotechnology 2020; 18:46. [PMID: 32169062 PMCID: PMC7071717 DOI: 10.1186/s12951-020-00606-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/09/2020] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Peripheral nerve injury is one common clinical disease worldwide, in which sciatic nerve is anatomically the most challenging to regenerate given its length and large cross-sectional area. For the present, autologous nerve grafting remains to be the most ideal strategy when treating with sciatic nerve injury. However, this method sacrifices healthy nerves and requires highly intensive surgery, still calling for other advanced alternatives for nerve grafting. RESULTS In this study, we utilized previously well-established gene delivery system to dually deliver plasmid DNA (pDNA) encoding vascular endothelial growth factor (VEGF) and nerve growth factor (NGF), exploring therapeutics for sciatic nerve injury. Low-molecular-weight branched polyethylenimine (bPEI) was constructed as the backbone structure of gene vectors, and it was further crosslinked to synthesize degradable polycations via the conjugation of dialdehydes. Potential synergistic effect between VEGF and NGF proteins were observed on rat sciatic nerve crush injury model in this study. CONCLUSIONS We concluded that dual delivery of plasmid VEGF and NGF as gene therapy could enhance sciatic nerve regeneration.
Collapse
Affiliation(s)
- Zhiwei Fang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.,Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China
| | - Xuemei Ge
- School of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Xuan Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Xu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yuanming Ouyang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China.
| |
Collapse
|