1
|
Schlidt K, Asgardoon M, Febre-Alemañy DA, El-Mallah JC, Waldron O, Dawes J, Agrawal S, Landmesser ME, Ravnic DJ. Surgical Bioengineering of the Microvasculature and Challenges in Clinical Translation. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40171780 DOI: 10.1089/ten.teb.2024.0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Tissue and organ dysfunction are major causes of worldwide morbidity and mortality with all medical specialties being impacted. Tissue engineering is an interdisciplinary field relying on the combination of scaffolds, cells, and biologically active molecules to restore form and function. However, clinical translation is still largely hampered by limitations in vascularization. Consequently, a thorough understanding of the microvasculature is warranted. This review provides an overview of (1) angiogenesis, including sprouting angiogenesis, intussusceptive angiogenesis, vascular remodeling, vascular co-option, and inosculation; (2) strategies for vascularized engineered tissue fabrication such as scaffold modulation, prevascularization, growth factor utilization, and cell-based approaches; (3) guided microvascular development via scaffold modulation with electromechanical cues, 3D bioprinting, and electrospinning; (4) surgical approaches to bridge the micro- and macrovasculatures in order to hasten perfusion; and (5) building specific vasculature in the context of tissue repair and organ transplantation, including skin, adipose, bone, liver, kidney, and lung. Our goal is to provide the reader with a translational overview that spans developmental biology, tissue engineering, and clinical surgery.
Collapse
Affiliation(s)
- Kevin Schlidt
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mohamadhossein Asgardoon
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - David A Febre-Alemañy
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Jessica C El-Mallah
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Olivia Waldron
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Jazzmyn Dawes
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Shailaja Agrawal
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mary E Landmesser
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Dino J Ravnic
- Zubar Plastic Surgery Research Laboratory, Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
2
|
Bell JA, Mayfield CK, Collon K, Chang S, Gallo MC, Lechtholz-Zey E, Ayad M, Sugiyam O, Tang AH, Park SH, Lieberman JR. In vivo effects of cell seeding technique in an ex vivo regional gene therapy model for bone regeneration. J Biomed Mater Res A 2024; 112:1688-1698. [PMID: 38602243 DOI: 10.1002/jbm.a.37718] [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: 10/04/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024]
Abstract
When delivering cells on a scaffold to treat a bone defect, the cell seeding technique determines the number and distribution of cells within a scaffold, however the optimal technique has not been established. This study investigated if human adipose-derived stem cells (ASCs) transduced with a lentiviral vector to overexpress bone morphogenetic protein 2 (BMP-2) and loaded on a scaffold using dynamic orbital shaker could reduce the total cell dose required to heal a critical sized bone defect when compared with static seeding. Human ASCs were loaded onto a collagen/biphasic ceramic scaffold using static loading and dynamic orbital shaker techniques, compared with our labs standard loading technique, and implanted into femoral defects of nude rats. Both a low dose and standard dose of transduced cells were evaluated. Outcomes investigated included BMP-2 production, radiographic healing, micro-computerized tomography, histologic assessment, and biomechanical torsional testing. BMP-2 production was higher in the orbital shaker cohort compared with the static seeding cohort. No statistically significant differences were noted in radiographic, histomorphometric, and biomechanical outcomes between the low-dose static and dynamic seeding groups, however the standard-dose static seeding cohort had superior biomechanical properties. The standard-dose 5 million cell dose standard loading cohort had superior maximum torque and torsional stiffness on biomechanical testing. The use of orbital shaker technique was labor intensive and did not provide equivalent biomechanical results with the use of fewer cells.
Collapse
Affiliation(s)
- Jennifer A Bell
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Cory K Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Kevin Collon
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Stephanie Chang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Matthew C Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Elizabeth Lechtholz-Zey
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Mina Ayad
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Osamu Sugiyam
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Sang-Hyun Park
- J. Vernon Luck Orthopaedic Research Center, Orthopaedic Institute for Children, Los Angeles, California, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| |
Collapse
|
3
|
Stoian A, Adil A, Biniazan F, Haykal S. Two Decades of Advances and Limitations in Organ Recellularization. Curr Issues Mol Biol 2024; 46:9179-9214. [PMID: 39194760 DOI: 10.3390/cimb46080543] [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: 07/31/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
The recellularization of tissues after decellularization is a relatively new technology in the field of tissue engineering (TE). Decellularization involves removing cells from a tissue or organ, leaving only the extracellular matrix (ECM). This can then be recellularized with new cells to create functional tissues or organs. The first significant mention of recellularization in decellularized tissues can be traced to research conducted in the early 2000s. One of the landmark studies in this field was published in 2008 by Ott, where researchers demonstrated the recellularization of a decellularized rat heart with cardiac cells, resulting in a functional organ capable of contraction. Since then, other important studies have been published. These studies paved the way for the widespread application of recellularization in TE, demonstrating the potential of decellularized ECM to serve as a scaffold for regenerating functional tissues. Thus, although the concept of recellularization was initially explored in previous decades, these studies from the 2000s marked a major turning point in the development and practical application of the technology for the recellularization of decellularized tissues. The article reviews the historical advances and limitations in organ recellularization in TE over the last two decades.
Collapse
Affiliation(s)
- Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Aisha Adil
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
- Reconstructive Oncology, Division of Plastic and Reconstructive Surgery, Smilow Cancer Hospital, Yale, New Haven, CT 06519, USA
| |
Collapse
|
4
|
Bedar M, Pulos NA, Shin AY. Dynamic Seeding versus Microinjection of Adipose-Derived Mesenchymal Stem Cells to Acellular Nerve Allograft Reconstructions. Plast Reconstr Surg 2024; 154:114e-125e. [PMID: 37537724 PMCID: PMC10838349 DOI: 10.1097/prs.0000000000010970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND Functional recovery after acellular nerve allograft (ANA) reconstruction remains inferior to that after autologous nerve grafting, but improved outcomes have been demonstrated with the addition of adipose-derived mesenchymal stem cells (MSCs). Controversy exists regarding the optimal cell-delivery method to enhance ANA reconstructions. The authors investigated the functional recovery of ANAs after dynamic seeding versus microinjection of MSCs. METHODS Forty Lewis rats underwent reconstruction of a 10-mm sciatic nerve defect. Animals were divided into 4 groups: reversed autograft, ANA alone, dynamically seeded ANA, or ANA injected with MSCs. During the survival period, ultrasound measurements of the tibialis anterior muscle cross-sectional area were performed. At 12 weeks, functional recovery was evaluated using measurements of ankle contracture, compound muscle action potential, maximum isometric tetanic force, muscle mass, histomorphometry, and immunofluorescence. RESULTS The dynamic seeding and microinjection groups demonstrated higher cross-sectional tibialis anterior muscle area recovery than autografts and ANAs alone at week 8 and weeks 4 and 8, respectively. The ankle contracture and compound muscle action potential amplitude recovery were superior in autografts and both seeding methods compared with ANAs alone. The microinjection group demonstrated significantly higher isometric tetanic force, muscle mass, and number of axons compared with ANAs alone. Both seeding methods showed higher CD34 densities compared with ANAs alone. No significant differences between dynamic seeding and microinjection were observed in functional or histologic outcomes. CONCLUSIONS The addition of MSCs to ANAs demonstrated earlier motor regeneration compared with autografts and ANAs alone. Both seeding methods improved functional outcomes in the rat sciatic nerve defect model.
Collapse
Affiliation(s)
- Meiwand Bedar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Plastic Surgery, Nijmegen, The Netherlands
| | | | | |
Collapse
|
5
|
Barcena AJR, Mishra A, Bolinas DKM, Martin BM, Melancon MP. Integration of Electrospun Scaffolds and Biological Polymers for Enhancing the Delivery and Efficacy of Mesenchymal Stem/Stromal Cell Therapies. FRONT BIOSCI-LANDMRK 2024; 29:228. [PMID: 38940050 PMCID: PMC11725061 DOI: 10.31083/j.fbl2906228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach for a variety of diseases due to their immunomodulatory and tissue regeneration capabilities. Despite their potential, the clinical application of MSC therapies is hindered by limited cell retention and engraftment at the target sites. Electrospun scaffolds, with their high surface area-to-volume ratio and tunable physicochemical properties, can be used as platforms for MSC delivery. However, synthetic polymers often lack the bioactive cues necessary for optimal cell-scaffold interactions. Integrating electrospun scaffolds and biological polymers, such as polysaccharides, proteins, and composites, combines the mechanical integrity of synthetic materials with the bioactivity of natural polymers and represents a strategic approach to enhance cell-scaffold interactions. The molecular interactions between MSCs and blended or functionalized scaffolds have been examined in recent studies, and it has been shown that integration can enhance MSC adhesion, proliferation, and paracrine secretion through the activation of multiple signaling pathways, such as FAK/Src, MAPK, PI3K/Akt, Wnt/β-catenin, and YAP/TAZ. Preclinical studies on small animals also reveal that the integration of electrospun scaffolds and natural polymers represents a promising approach to enhancing the delivery and efficacy of MSCs in the context of regenerating bone, cartilage, muscle, cardiac, vascular, and nervous tissues. Future research should concentrate on identifying the distinct characteristics of the MSC niche, investigating the processes involved in MSC-scaffold interactions, and applying new technologies in stem cell treatment and biofabrication to enhance scaffold design. Research on large animal models and collaboration among materials scientists, engineers, and physicians are crucial to translating these advancements into clinical use.
Collapse
Affiliation(s)
- Allan John R. Barcena
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Archana Mishra
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dominic Karl M. Bolinas
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Benjamin M. Martin
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
6
|
Jeang WJ, Bochenek MA, Bose S, Zhao Y, Wong BM, Yang J, Jiang AL, Langer R, Anderson DG. Silicone cryogel skeletons enhance the survival and mechanical integrity of hydrogel-encapsulated cell therapies. SCIENCE ADVANCES 2024; 10:eadk5949. [PMID: 38578991 PMCID: PMC10997197 DOI: 10.1126/sciadv.adk5949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 03/01/2024] [Indexed: 04/07/2024]
Abstract
The transplantation of engineered cells that secrete therapeutic proteins presents a promising method for addressing a range of chronic diseases. However, hydrogels used to encase and protect non-autologous cells from immune rejection often suffer from poor mechanical properties, insufficient oxygenation, and fibrotic encapsulation. Here, we introduce a composite encapsulation system comprising an oxygen-permeable silicone cryogel skeleton, a hydrogel matrix, and a fibrosis-resistant polymer coating. Cryogel skeletons enhance the fracture toughness of conventional alginate hydrogels by 23-fold and oxygen diffusion by 2.8-fold, effectively mitigating both implant fracture and hypoxia of encapsulated cells. Composite implants containing xenogeneic cells engineered to secrete erythropoietin significantly outperform unsupported alginate implants in therapeutic delivery over 8 weeks in immunocompetent mice. By improving mechanical resiliency and sustaining denser cell populations, silicone cryogel skeletons enable more durable and miniaturized therapeutic implants.
Collapse
Affiliation(s)
- William J. Jeang
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Matthew A. Bochenek
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Suman Bose
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Yichao Zhao
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bryan M. Wong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jiawei Yang
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Alexis L. Jiang
- Department of Computer Science, Wellesley College, Wellesley, MA 02481, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G. Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
7
|
Masri S, Fadilah NIM, Hao LQ, Maarof M, Tabata Y, Hiraoka Y, Fauzi MB. Multifunctionalised skin substitute of hybrid gelatin-polyvinyl alcohol bioinks for chronic wound: injectable vs. 3D bioprinting. Drug Deliv Transl Res 2024; 14:1005-1027. [PMID: 37938542 DOI: 10.1007/s13346-023-01447-z] [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] [Accepted: 10/06/2023] [Indexed: 11/09/2023]
Abstract
Chronic wounds are challenging to heal and increase global mortality. The effectiveness of skin graft is limited by rejection, fibrosis, and inadequate donor site. Multifunctionalised-hydrogel skin substitutes promoted higher wound healing by maintaining the moisture microenvironment and permit gas exchange/nourishment in prolong cell viability/activity. The purpose of this study was to evaluate a skin substitute using two strategies; via injectable and 3D bioprinting technique. New hydrogel formulations that composed of gelatin (GE) and polyvinyl-alcohol (PVA) were constructed using a pre-mix crosslinking approach with genipin (GNP) to generate the biodegradable and biocompatible skin substitute with reduced secondary traumatic wound. GPVA5_GNP (6% GE: 5% PVA crosslinked with GNP) was the most stable hydrogel for wound healing application with the longest enzymatic degradation and stable hydrogel for absorption of excess wound exudates. Primary human dermal fibroblasts (HDFs) migrated extensively through 3D bioprinted hydrogels with larger average pore sizes and interconnected pores than injectable hydrogels. Moreover, 3D bioprinted GPVA hydrogels were biocompatible with HDFs and demonstrated > 90% cell viability. HDFs maintained their phenotype and positively expressed collagen type-I, vinculin, short and dense F-actin, alpha-smooth muscle actin, and Ki67. Additionally, the presence of GNP demonstrated antioxidant capacity and high-ability of angiogenesis. The utilisation of the 3D bioprinting (layer-by-layer) approach did not compromise the HDFs' growth capacity and biocompatibility with selected bioinks. In conclusion, it allows the cell encapsulation sustainability in a hydrogel matrix for a longer period, in promoting tissue regeneration and accelerating healing capacity, especially for difficult or chronic wound.
Collapse
Affiliation(s)
- Syafira Masri
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, 15th Floor Pre-Clinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Nur Izzah Md Fadilah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, 15th Floor Pre-Clinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Looi Qi Hao
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, 15th Floor Pre-Clinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
- My Cytohealth Sdn. Bhd, 56000, Kuala Lumpur, Malaysia
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, 15th Floor Pre-Clinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Science (LiMe), Kyoto University, Kyoto, 606-8500, Japan
| | - Yosuke Hiraoka
- Biomaterial Group, R&D Center, Yao City, 581-0000, Japan
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, 15th Floor Pre-Clinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia.
- My Cytohealth Sdn. Bhd, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
8
|
Mbitta Akoa D, Sicard L, Hélary C, Torrens C, Baroukh B, Poliard A, Coradin T. Role of Physico-Chemical and Cellular Conditions on the Bone Repair Potential of Plastically Compressed Collagen Hydrogels. Gels 2024; 10:130. [PMID: 38391460 PMCID: PMC10887598 DOI: 10.3390/gels10020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Since their first description nearly 20 years ago, dense collagen hydrogels obtained by plastic compression have become popular scaffolds in tissue engineering. In particular, when seeded with dental pulp stem cells, they have demonstrated a great in vivo potential in cranial bone repair. Here, we investigated how physico-chemical and cell-seeding conditions could influence the formation and in vitro mineralization of these cellularized scaffolds. A qualitative assessment demonstrated that the gel stability before and after compression was highly sensitive to the conditions of fibrillogenesis, especially initial acid acetic and buffer concentrations. Gels with similar rheological properties but different fibrillar structures that exhibited different stabilities when used for the 3D culture of Stem cells from Human Exfoliated Deciduous teeth (SHEDs) could be prepared. Finally, in our optimal physico-chemical conditions, mineralization could be achieved only using human dental pulp stem cells (hDPSCs) at a high cell density. These results highlight the key role of fibrillogenic conditions and cell type/density on the bone repair potential of cell-laden plastically compressed collagen hydrogels.
Collapse
Affiliation(s)
- Daline Mbitta Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Ludovic Sicard
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
- AP-HP Service de Médecine Bucco-Dentaire, Hôpital Bretonneau, 75018 Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Coralie Torrens
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
| | - Brigitte Baroukh
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
| | - Anne Poliard
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| |
Collapse
|
9
|
Carrabba M, Fagnano M, Ghorbel MT. Development of a Novel Hierarchically Biofabricated Blood Vessel Mimic Decorated with Three Vascular Cell Populations for the Reconstruction of Small-Diameter Arteries. ADVANCED FUNCTIONAL MATERIALS 2024; 34:adfm.202300621. [PMID: 39257639 PMCID: PMC7616429 DOI: 10.1002/adfm.202300621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Indexed: 09/12/2024]
Abstract
The availability of grafts to replace small-diameter arteries remains an unmet clinical need. Here, the validated methodology is reported for a novel hybrid tissue-engineered vascular graft that aims to match the natural structure of small-size arteries. The blood vessel mimic (BVM) comprises an internal conduit of co-electrospun gelatin and polycaprolactone (PCL) nanofibers (corresponding to the tunica intima of an artery), reinforced by an additional layer of PCL aligned fibers (the internal elastic membrane). Endothelial cells are deposited onto the luminal surface using a rotative bioreactor. A bioprinting system extrudes two concentric cell-laden hydrogel layers containing respectively vascular smooth muscle cells and pericytes to create the tunica media and adventitia. The semi-automated cellularization process reduces the production and maturation time to 6 days. After the evaluation of mechanical properties, cellular viability, hemocompatibility, and suturability, the BVM is successfully implanted in the left pulmonary artery of swine. Here, the BVM showed good hemostatic properties, capability to withstand blood pressure, and patency at 5 weeks post-implantation. These promising data open a new avenue to developing an artery-like product for reconstructing small-diameter blood vessels.
Collapse
Affiliation(s)
- Michele Carrabba
- Bristol Heart Institute, School of Translational Health Sciences, Bristol Medical School, University of Bristol, BristolBS2 8HW, UK
| | - Marco Fagnano
- Bristol Heart Institute, School of Translational Health Sciences, Bristol Medical School, University of Bristol, BristolBS2 8HW, UK
| | - Mohamed T Ghorbel
- Bristol Heart Institute, School of Translational Health Sciences, Bristol Medical School, University of Bristol, BristolBS2 8HW, UK
| |
Collapse
|
10
|
Jeong HJ, Nam H, Kim JS, Cho S, Park HH, Cho YS, Jeon H, Jang J, Lee SJ. Dragging 3D printing technique controls pore sizes of tissue engineered blood vessels to induce spontaneous cellular assembly. Bioact Mater 2024; 31:590-602. [PMID: 37876874 PMCID: PMC10593581 DOI: 10.1016/j.bioactmat.2023.07.021] [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: 02/09/2023] [Revised: 06/23/2023] [Accepted: 07/24/2023] [Indexed: 10/26/2023] Open
Abstract
To date, several off-the-shelf products such as artificial blood vessel grafts have been reported and clinically tested for small diameter vessel (SDV) replacement. However, conventional artificial blood vessel grafts lack endothelium and, thus, are not ideal for SDV transplantation as they can cause thrombosis. In addition, a successful artificial blood vessel graft for SDV must have sufficient mechanical properties to withstand various external stresses. Here, we developed a spontaneous cellular assembly SDV (S-SDV) that develops without additional intervention. By improving the dragging 3D printing technique, SDV constructs with free-form, multilayers and controllable pore size can be fabricated at once. Then, The S-SDV filled in the natural polymer bioink containing human umbilical vein endothelial cells (HUVECs) and human aorta smooth muscle cells (HAoSMCs). The endothelium can be induced by migration and self-assembly of endothelial cells through pores of the SDV construct. The antiplatelet adhesion of the formed endothelium on the luminal surface was also confirmed. In addition, this S-SDV had sufficient mechanical properties (burst pressure, suture retention, leakage test) for transplantation. We believe that the S-SDV could address the challenges of conventional SDVs: notably, endothelial formation and mechanical properties. In particular, the S-SDV can be designed simply as a free-form structure with a desired pore size. Since endothelial formation through the pore is easy even in free-form constructs, it is expected to be useful for endothelial formation in vascular structures with branch or curve shapes, and in other tubular tissues such as the esophagus.
Collapse
Affiliation(s)
- Hun-Jin Jeong
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
- Regenerative Engineering Laboratory, Columbia University, 630W 168th ST, New York, 10032, USA
| | - Hyoryung Nam
- Department of Convergence IT Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
| | - Jae-Seok Kim
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Sungkeon Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
| | - Hyun-Ha Park
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Young-Sam Cho
- Department of Mechanical and Design Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Hyungkook Jeon
- Department of Manufacturing Systems and Design Engineering, Seoul National University of Science and Technology, 01811, Seoul, Republic of Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 37673, Pohang, Gyeongbuk, Republic of Korea
- Institute of Convergence Science, Yonsei University, 03722, Seoul, Republic of Korea
| | - Seung-Jae Lee
- Department of Mechanical and Design Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| |
Collapse
|
11
|
Snyder Y, Jana S. Strategies for Development of Synthetic Heart Valve Tissue Engineering Scaffolds. PROGRESS IN MATERIALS SCIENCE 2023; 139:101173. [PMID: 37981978 PMCID: PMC10655624 DOI: 10.1016/j.pmatsci.2023.101173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
The current clinical solutions, including mechanical and bioprosthetic valves for valvular heart diseases, are plagued by coagulation, calcification, nondurability, and the inability to grow with patients. The tissue engineering approach attempts to resolve these shortcomings by producing heart valve scaffolds that may deliver patients a life-long solution. Heart valve scaffolds serve as a three-dimensional support structure made of biocompatible materials that provide adequate porosity for cell infiltration, and nutrient and waste transport, sponsor cell adhesion, proliferation, and differentiation, and allow for extracellular matrix production that together contributes to the generation of functional neotissue. The foundation of successful heart valve tissue engineering is replicating native heart valve architecture, mechanics, and cellular attributes through appropriate biomaterials and scaffold designs. This article reviews biomaterials, the fabrication of heart valve scaffolds, and their in-vitro and in-vivo evaluations applied for heart valve tissue engineering.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA
| | - Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
12
|
Wang X, Elbahrawi RT, Abdukadir AM, Ali ZM, Chan V, Corridon PR. A proposed model of xeno-keratoplasty using 3D printing and decellularization. Front Pharmacol 2023; 14:1193606. [PMID: 37799970 PMCID: PMC10548234 DOI: 10.3389/fphar.2023.1193606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 09/06/2023] [Indexed: 10/07/2023] Open
Abstract
Corneal opacity is a leading cause of vision impairment and suffering worldwide. Transplantation can effectively restore vision and reduce chronic discomfort. However, there is a considerable shortage of viable corneal graft tissues. Tissue engineering may address this issue by advancing xeno-keratoplasty as a viable alternative to conventional keratoplasty. In particular, livestock decellularization strategies offer the potential to generate bioartificial ocular prosthetics in sufficient supply to match existing and projected needs. To this end, we have examined the best practices and characterizations that have supported the current state-of-the-art driving preclinical and clinical applications. Identifying the challenges that delimit activities to supplement the donor corneal pool derived from acellular scaffolds allowed us to hypothesize a model for keratoprosthesis applications derived from livestock combining 3D printing and decellularization.
Collapse
Affiliation(s)
- Xinyu Wang
- Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Rawdah Taha Elbahrawi
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Azhar Mohamud Abdukadir
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Zehara Mohammed Ali
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Vincent Chan
- Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter R. Corridon
- Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- Hleathcare, Engineering and Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Johnston A, Callanan A. Recent Methods for Modifying Mechanical Properties of Tissue-Engineered Scaffolds for Clinical Applications. Biomimetics (Basel) 2023; 8:205. [PMID: 37218791 PMCID: PMC10204517 DOI: 10.3390/biomimetics8020205] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
The limited regenerative capacity of the human body, in conjunction with a shortage of healthy autologous tissue, has created an urgent need for alternative grafting materials. A potential solution is a tissue-engineered graft, a construct which supports and integrates with host tissue. One of the key challenges in fabricating a tissue-engineered graft is achieving mechanical compatibility with the graft site; a disparity in these properties can shape the behaviour of the surrounding native tissue, contributing to the likelihood of graft failure. The purpose of this review is to examine the means by which researchers have altered the mechanical properties of tissue-engineered constructs via hybrid material usage, multi-layer scaffold designs, and surface modifications. A subset of these studies which has investigated the function of their constructs in vivo is also presented, followed by an examination of various tissue-engineered designs which have been clinically translated.
Collapse
Affiliation(s)
| | - Anthony Callanan
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh EH9 3DW, UK;
| |
Collapse
|
14
|
Guo C, Zhang T, Tang J, Gao C, Zhou Z, Li C. Construction of PLGA Porous Microsphere-Based Artificial Pancreatic Islets Assisted by the Cell Centrifugation Perfusion Technique. ACS OMEGA 2023; 8:15288-15297. [PMID: 37151553 PMCID: PMC10157690 DOI: 10.1021/acsomega.3c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023]
Abstract
Pancreatic islet transplantation is a promising treatment that could potentially reverse diabetes, but its clinical applicability is severely limited by a shortage of organ donors. Various cell loading approaches using polymeric porous microspheres (PMs) have been developed for tissue regeneration; however, PM-based multicellular artificial pancreatic islets' construction has been scarcely reported. In this study, MIN6 (a mouse insulinoma cell line) and MS1 (a mouse pancreatic islet endothelial cell line) cells were seeded into poly(lactic-co-glycolic acid) (PLGA) PMs via an upgraded centrifugation-based cell perfusion seeding technique invented and patented by our group. Cell morphology, distribution, viability, migration, and proliferation were all evaluated. Results from glucose-stimulated insulin secretion (GSIS) assay and RNA-seq analysis suggested that MIN6 and MS1-loaded PLGA PMs exhibited better glucose responsiveness, which is partly attributable to vascular formation during PM-dependent islet construction. The present study suggests that the PLGA PM-based artificial pancreatic islets may provide an alternative strategy for the potential treatment of diabetes in the future.
Collapse
Affiliation(s)
- Chuanjia Guo
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Tong Zhang
- Clinical
Laboratory, Tianjin Hospital, Tianjin 300211, China
| | - Jianghai Tang
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Chang Gao
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Zhimin Zhou
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
- ,
| | - Chen Li
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| |
Collapse
|
15
|
Nguyen HT, Peirsman A, Tirpakova Z, Mandal K, Vanlauwe F, Maity S, Kawakita S, Khorsandi D, Herculano R, Umemura C, Yilgor C, Bell R, Hanson A, Li S, Nanda HS, Zhu Y, Najafabadi AH, Jucaud V, Barros N, Dokmeci MR, Khademhosseini A. Engineered Vasculature for Cancer Research and Regenerative Medicine. MICROMACHINES 2023; 14:978. [PMID: 37241602 PMCID: PMC10221678 DOI: 10.3390/mi14050978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
Engineered human tissues created by three-dimensional cell culture of human cells in a hydrogel are becoming emerging model systems for cancer drug discovery and regenerative medicine. Complex functional engineered tissues can also assist in the regeneration, repair, or replacement of human tissues. However, one of the main hurdles for tissue engineering, three-dimensional cell culture, and regenerative medicine is the capability of delivering nutrients and oxygen to cells through the vasculatures. Several studies have investigated different strategies to create a functional vascular system in engineered tissues and organ-on-a-chips. Engineered vasculatures have been used for the studies of angiogenesis, vasculogenesis, as well as drug and cell transports across the endothelium. Moreover, vascular engineering allows the creation of large functional vascular conduits for regenerative medicine purposes. However, there are still many challenges in the creation of vascularized tissue constructs and their biological applications. This review will summarize the latest efforts to create vasculatures and vascularized tissues for cancer research and regenerative medicine.
Collapse
Affiliation(s)
- Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Zuzana Tirpakova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Florian Vanlauwe
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Rondinelli Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Bioengineering & Biomaterials Group, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil
| | - Christian Umemura
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Adrian Hanson
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Himansu Sekhar Nanda
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Biomedical Engineering and Technology Laboratory, PDPM—Indian Institute of Information Technology Design Manufacturing, Jabalpur 482005, Madhya Pradesh, India
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Natan Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| |
Collapse
|
16
|
Abuarqoub D, Theeb LS, Omari MB, Hamadneh YI, Alrawabdeh JA, Aslam N, Jafar H, Awidi A. The Osteogenic Role of Biomaterials Combined with Human-Derived Dental Stem Cells in Bone Tissue Regeneration. Tissue Eng Regen Med 2023; 20:251-270. [PMID: 36808303 PMCID: PMC10070593 DOI: 10.1007/s13770-022-00514-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 02/23/2023] Open
Abstract
The use of stem cells in regenerative medicine had great potential for clinical applications. However, cell delivery strategies have critical importance in stimulating the differentiation of stem cells and enhancing their potential to regenerate damaged tissues. Different strategies have been used to investigate the osteogenic potential of dental stem cells in conjunction with biomaterials through in vitro and in vivo studies. Osteogenesis has a broad implication in regenerative medicine, particularly for maxillofacial defects. This review summarizes some of the most recent developments in the field of tissue engineering using dental stem cells.
Collapse
Affiliation(s)
- Duaa Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
| | - Laith S Theeb
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Mohammad B Omari
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Yazan I Hamadneh
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | | | - Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
- School of Medicine, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
17
|
Nun N, Joy A. Fabrication and Bioactivity of Peptide-Conjugated Biomaterial Tissue Engineering Constructs. Macromol Rapid Commun 2023; 44:e2200342. [PMID: 35822458 DOI: 10.1002/marc.202200342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/22/2022] [Indexed: 01/11/2023]
Abstract
Tissue engineering combines materials engineering, cells and biochemical factors to improve, restore or replace various types of biological tissues. A nearly limitless combination of these strategies can be combined, providing a means to augment the function of a number of biological tissues such as skin tissue, neural tissue, bones, and cartilage. Compounds such as small molecule therapeutics, proteins, and even living cells have been incorporated into tissue engineering constructs to influence biological processes at the site of implantation. Peptides have been conjugated to tissue engineering constructs to circumvent limitations associated with conjugation of proteins or incorporation of cells. This review highlights various contemporary examples in which peptide conjugation is used to overcome the disadvantages associated with the inclusion of other bioactive compounds. This review covers several peptides that are commonly used in the literature as well as those that do not appear as frequently to provide a broad scope of the utility of the peptide conjugation technique for designing constructs capable of influencing the repair and regeneration of various bodily tissues. Additionally, a brief description of the construct fabrication techniques encountered in the covered examples and their advantages in various tissue engineering applications is provided.
Collapse
Affiliation(s)
- Nicholas Nun
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| | - Abraham Joy
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| |
Collapse
|
18
|
Collon K, Bell JA, Chang SW, Gallo MC, Sugiyama O, Marks C, Lieberman JR. Effects of cell seeding technique and cell density on BMP-2 production in transduced human mesenchymal stem cells. J Biomed Mater Res A 2022; 110:1944-1952. [PMID: 35950648 PMCID: PMC9804863 DOI: 10.1002/jbm.a.37430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 01/09/2023]
Abstract
Small animal models have demonstrated the efficacy of ex vivo regional gene therapy using scaffolds loaded with BMP-2-expressing mesenchymal stem cells (MSCs). Prior to clinical translation, optimization of seeding techniques of the transduced cells will be important to minimize time and resource expenditure, while maximizing cell delivery and BMP-2 production. No prior studies have investigated cell-seeding techniques in the setting of transduced cells for gene therapy applications. Using BMP-2-expressing transduced adipose-derived MSCs and a porous ceramic scaffold, this study compared previously described static and dynamic seeding techniques with respect to cell seeding efficiency, uniformity of cell distribution, and in vitro BMP-2 production. Static and negative pressure seeding techniques demonstrated the highest seeding efficiency, while orbital shaking was associated with the greatest increases in BMP-2 production per cell. Low density cell suspensions were associated with the highest seeding efficiency and uniformity of cell distribution, and the greatest increases in BMP-2 production from 2 to 7 days after seeding. Our results highlight the potential for development of an optimized cell density and seeding technique that could greatly reduce the number of MSCs needed to produce therapeutic BMP-2 levels in clinical situations. Further studies are needed to investigate in vivo effects of cell seeding techniques on bone healing.
Collapse
Affiliation(s)
- Kevin Collon
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Jennifer A. Bell
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Stephanie W. Chang
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Matthew C. Gallo
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Osamu Sugiyama
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Carolyn Marks
- Core Center of Excellence in Nano ImagingUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jay R. Lieberman
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| |
Collapse
|
19
|
Amagat J, Su Y, Svejsø FH, Le Friec A, Sønderskov SM, Dong M, Fang Y, Chen M. Self-snapping hydrogel-based electroactive microchannels as nerve guidance conduits. Mater Today Bio 2022; 16:100437. [PMID: 36193343 PMCID: PMC9526217 DOI: 10.1016/j.mtbio.2022.100437] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/01/2022] Open
Abstract
Peripheral nerve regeneration with large defects needs innovative design of nerve guidance conduits (NGCs) which possess anisotropic guidance, electrical induction and right mechanical properties in one. Herein, we present, for the first time, facile fabrication and efficient neural differentiation guidance of anisotropic, conductive, self-snapping, hydrogel-based NGCs. The hydrogels were fabricated via crosslinking of graphitic carbon nitride (g-C3N4) upon exposure with blue light, incorporated with graphene oxide (GO). Incorporation of GO and in situ reduction greatly enhanced surface charges, while decayed light penetration endowed the hydrogel with an intriguing self-snapping feature by the virtue of a crosslinking gradient. The hydrogels were in the optimal mechanical stiffness range for peripheral nerve regeneration and supported normal viability and proliferation of neural cells. The PC12 cells differentiated on the electroactive g-C3N4 H/rGO3 (3 mg/mL GO loading) hydrogel presented 47% longer neurite length than that of the pristine g-C3N4 H hydrogel. Furthermore, the NGC with aligned microchannels was successfully fabricated using sacrificial melt electrowriting (MEW) moulding, the anisotropic microchannels of the 10 μm width showed optimal neurite guidance. Such anisotropic, electroactive, self-snapping NGCs may possess great potential for repairing peripheral nerve injuries.
Collapse
|
20
|
García-Sobrino R, Lago E, Goñi C, Ramos V, García C, Reinecke H, Elvira C, Rodríguez-Hernández J, Gallardo A, Martínez-Campos E. Fabrication of 3D cylindrical thermosensitive hydrogels as supports for cell culture and detachment of tubular cell sheets. BIOMATERIALS ADVANCES 2022; 144:213210. [PMID: 36473351 DOI: 10.1016/j.bioadv.2022.213210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/31/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Pseudo interpenetrating vinyl-caprolactam (VCL) based thermosensitive tubular hydrogels with a volume phase transition temperature, VPTT, around 35 °C, have been prepared by combining two different crosslinkers, a di-methacrylate (C1) and a di-vinyl urea (C2). The molar ratio between the two crosslinkers (for a global crosslinker molar percentage of 1.9) has shown to play a key role on the properties of the hydrogel. Increasing the amount of di-vinyl urea, leads to transparent but rather fragile materials and to a lower extent of thermosensitivity, that is, to a lower variation in the hydrogel swelling upon temperature change. However, tubes prepared with a selected crosslinker molar ratio C1/C2 of 65/35 provided a compromise between transparency, thermosensitivity and maneuverability and were, thus, evaluated as supports for cell culture using premyoblastic cells. These hydrogels, used as supports, allow for surface adhesion and cell proliferation until confluence, and eventually an efficient monolayer detachment (and transplant to a 3D-printed polylactic acid (PLA) support) through a controlled drop in temperature. As a result, this method permits to obtain tubular tissue constructs with potential applications in tissue engineering such as in the elaboration of vascular grafts.
Collapse
Affiliation(s)
- Rubén García-Sobrino
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain; Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid, Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
| | - Eugenia Lago
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Clara Goñi
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Viviana Ramos
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid, Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
| | - Carolina García
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Helmut Reinecke
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Carlos Elvira
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Juan Rodríguez-Hernández
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Alberto Gallardo
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Enrique Martínez-Campos
- Polymer Functionalization Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Departamento de Química Macromolecular Aplicada, Juan de la Cierva 3, 28006 Madrid, Spain; Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid, Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain.
| |
Collapse
|
21
|
Lindner M, Laporte A, Elomaa L, Lee-Thedieck C, Olmer R, Weinhart M. Flow-induced glycocalyx formation and cell alignment of HUVECs compared to iPSC-derived ECs for tissue engineering applications. Front Cell Dev Biol 2022; 10:953062. [PMID: 36133919 PMCID: PMC9483120 DOI: 10.3389/fcell.2022.953062] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/19/2022] [Indexed: 12/03/2022] Open
Abstract
The relevance of cellular in vitro models highly depends on their ability to mimic the physiological environment of the respective tissue or cell niche. Static culture conditions are often unsuitable, especially for endothelial models, since they completely neglect the physiological surface shear stress and corresponding reactions of endothelial cells (ECs) such as alignment in the direction of flow. Furthermore, formation and maturation of the glycocalyx, the essential polysaccharide layer covering all endothelial surfaces and regulating diverse processes, is highly dependent on applied fluid flow. This fragile but utterly important macromolecular layer is hard to analyze, its importance is often underestimated and accordingly neglected in many endothelial models. Therefore, we exposed human umbilical vein ECs (HUVECs) and human induced pluripotent stem cell-derived ECs (iPSC-ECs) as two relevant EC models in a side-by-side comparison to static and physiological dynamic (6.6 dyn cm-2) culture conditions. Both cell types demonstrated an elongation and alignment along the flow direction, some distinct changes in glycocalyx composition on the surface regarding the main glycosaminoglycan components heparan sulfate, chondroitin sulfate or hyaluronic acid as well as an increased and thereby improved glycocalyx thickness and functionality when cultured under homogeneous fluid flow. Thus, we were able to demonstrate the maturity of the employed iPSC-EC model regarding its ability to sense fluid flow along with the general importance of physiological shear stress for glycocalyx formation. Additionally, we investigated EC monolayer integrity with and without application of surface shear stress, revealing a comparable existence of tight junctions for all conditions and a reorganization of the cytoskeleton upon dynamic culture leading to an increased formation of focal adhesions. We then fabricated cell sheets of EC monolayers after static and dynamic culture via non-enzymatic detachment using thermoresponsive polymer coatings as culture substrates. In a first proof-of-concept we were able to transfer an aligned iPSC-EC sheet to a 3D-printed scaffold thereby making a step in the direction of vascular modelling. We envision these results to be a valuable contribution to improvements of in vitro endothelial models and vascular engineering in the future.
Collapse
Affiliation(s)
- Marcus Lindner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Anna Laporte
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Cornelia Lee-Thedieck
- Institute of Cell Biology and Biophysics, Leibniz Universität Hannover, Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- REBIRTH–Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
| |
Collapse
|
22
|
Tissue Engineering Approaches to Uncover Therapeutic Targets for Endothelial Dysfunction in Pathological Microenvironments. Int J Mol Sci 2022; 23:ijms23137416. [PMID: 35806421 PMCID: PMC9266895 DOI: 10.3390/ijms23137416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023] Open
Abstract
Endothelial cell dysfunction plays a central role in many pathologies, rendering it crucial to understand the underlying mechanism for potential therapeutics. Tissue engineering offers opportunities for in vitro studies of endothelial dysfunction in pathological mimicry environments. Here, we begin by analyzing hydrogel biomaterials as a platform for understanding the roles of the extracellular matrix and hypoxia in vascular formation. We next examine how three-dimensional bioprinting has been applied to recapitulate healthy and diseased tissue constructs in a highly controllable and patient-specific manner. Similarly, studies have utilized organs-on-a-chip technology to understand endothelial dysfunction's contribution to pathologies in tissue-specific cellular components under well-controlled physicochemical cues. Finally, we consider studies using the in vitro construction of multicellular blood vessels, termed tissue-engineered blood vessels, and the spontaneous assembly of microvascular networks in organoids to delineate pathological endothelial dysfunction.
Collapse
|
23
|
Strategies for development of decellularized heart valve scaffolds for tissue engineering. Biomaterials 2022; 288:121675. [DOI: 10.1016/j.biomaterials.2022.121675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 01/01/2023]
|
24
|
Saunders SK, Cole SY, Acuna Sierra V, Bracamonte JH, Toldo S, Soares JS. Evaluation of perfusion-driven cell seeding of small diameter engineered tissue vascular grafts with a custom-designed seed-and-culture bioreactor. PLoS One 2022; 17:e0269499. [PMID: 35709083 PMCID: PMC9202848 DOI: 10.1371/journal.pone.0269499] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/20/2022] [Indexed: 11/24/2022] Open
Abstract
Tissue engineering commonly entails combining autologous cell sources with biocompatible scaffolds for the replacement of damaged tissues in the body. Scaffolds provide functional support while also providing an ideal environment for the growth of new tissues until host integration is complete. To expedite tissue development, cells need to be distributed evenly within the scaffold. For scaffolds with a small diameter tubular geometry, like those used for vascular tissue engineering, seeding cells evenly along the luminal surface can be especially challenging. Perfusion-based cell seeding methods have been shown to promote increased uniformity in initial cell distribution onto porous scaffolds for a variety of tissue engineering applications. We investigate the seeding efficiency of a custom-designed perfusion-based seed-and-culture bioreactor through comparisons to a static injection counterpart method and a more traditional drip seeding method. Murine vascular smooth muscle cells were seeded onto porous tubular electrospun polycaprolactone scaffolds, 2 mm in diameter and 30 mm in length, using the three methods, and allowed to rest for 24 hours. Once harvested, scaffolds were evaluated longitudinally and circumferentially to assess the presence of viable cells using alamarBlue and live/dead cell assays and their distribution with immunohistochemistry and scanning electron microscopy. On average, bioreactor-mediated perfusion seeding achieved 35% more luminal surface coverage when compared to static methods. Viability assessment demonstrated that the total number of viable cells achieved across methods was comparable with slight advantage to the bioreactor-mediated perfusion-seeding method. The method described is a simple, low-cost method to consistently obtain even distribution of seeded cells onto the luminal surfaces of small diameter tubular scaffolds.
Collapse
Affiliation(s)
- Sarah K. Saunders
- Department of Mechanical and Nuclear Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sam Y. Cole
- Department of Mechanical and Nuclear Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Valeria Acuna Sierra
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Johane H. Bracamonte
- Department of Mechanical and Nuclear Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Stefano Toldo
- Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Joao S. Soares
- Department of Mechanical and Nuclear Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
25
|
Zivari-Ghader T, Dolati S, Mehdizadeh A, Davaran S, Rashidi MR, Yousefi M. Recent scaffold-based tissue engineering approaches in premature ovarian failure treatment. J Tissue Eng Regen Med 2022; 16:605-620. [PMID: 35511799 DOI: 10.1002/term.3306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022]
Abstract
Recently, tissue engineering and regenerative medicine have received significant attention with outstanding advances. The main scope of this technology is to recover the damaged tissues and organs or to maintain and improve their function. One of the essential fields in tissue engineering is scaffold designing and construction, playing an integral role in damaged tissues reconstruction and repair. However, premature ovarian failure (POF) is a disorder causing many medical and psychological problems in women. POF treatment using tissue engineering and various scaffold has recently made tremendous and promising progress. Due to the importance of the subject, we have summarized the recently examined scaffolds in the treatment of POF in this review.
Collapse
Affiliation(s)
- Tayyebeh Zivari-Ghader
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Dynamic seeding versus microinjection of mesenchymal stem cells for acellular nerve allograft: an in vitro comparison. J Plast Reconstr Aesthet Surg 2022; 75:2821-2830. [PMID: 35570113 PMCID: PMC9391259 DOI: 10.1016/j.bjps.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/17/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-supplemented acellular nerve allografts (ANA) are a potential strategy to improve the treatment of segmental nerve defects. Prior to clinical translation, optimal cell delivery methods must be defined. While two techniques, dynamic seeding and microinjection, have been described, the seeding efficiency, cell viability, and distribution of MSCs in ANAs are yet to be compared. METHODS Sciatic nerve segments of Sprague-Dawley rats were decellularized, and MSCs were harvested from the adipose tissue of Lewis rats. Cell viability was evaluated after injection of MSCs through a 27-gauge needle at different flow rates (10, 5, and 1 µL/min). MSCs were dynamically seeded or longitudinally injected into ANAs. Cell viability, seeding efficiency, and distribution were evaluated using LIVE/DEAD and MTS assays, scanning electron microscopy, and Hoechst staining. RESULTS No statistically significant difference in cell viability after injection at different flow rates was seen. After cell delivery, 84.1 ± 3.7% and 87.8 ± 2.8% of MSCs remained viable in the dynamic seeding and microinjection group, respectively (p = 0.41). The seeding efficiency of microinjection (100.4%±5.6) was significantly higher than dynamic seeding (48.1%±8.6) on day 1 (p = 0.001). Dynamic seeding demonstrated a significantly more uniform cell distribution over the course of the ANA compared to microinjection (p = 0.02). CONCLUSION MSCs remain viable after both dynamic seeding and microinjection in ANAs. Higher seeding efficiency was observed with microinjection, but dynamic seeding resulted in a more uniform distribution. In vivo studies are required to assess the effect on gene expression profiles and functional motor outcomes.
Collapse
|
27
|
Michael P, Yang N, Moore M, Santos M, Lam YT, Ward A, Hung JC, Tan R, Wise S. Synthetic Vascular Graft with Spatially Distinct Architecture for Rapid Biomimetic Cell Organisation in a Perfusion Bioreactor. Biomed Mater 2022; 17. [PMID: 35413704 DOI: 10.1088/1748-605x/ac66b2] [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: 12/10/2021] [Accepted: 04/12/2022] [Indexed: 11/12/2022]
Abstract
Access to lab-grown fully functional blood vessels would provide an invaluable resource to vascular medicine. The complex architecture and cellular makeup of native vessels, however, makes this extremely challenging to reproduce in vitro. Bioreactor systems have helped advanced research in this area by replicating many of the physiological conditions necessary for full-scale tissue growth outside of the body. A key element underpinning these technologies are 3D vascular graft templates which serve as temporary scaffolds to direct cell growth into similar cellular architectures observed in native vessels. Grafts further engineered with appropriate physical cues to accommodate the multiple cell types that reside within native vessels may help improve the production efficiency and physiological accuracy of bioreactor-grown vessel substitutes. Here, we engineered two distinct scaffold architectures into an electrospun vascular graft aiming to encourage the spatial organisation of human vascular endothelial cells (hCAECs) in a continuous luminal monolayer, co-cultured with human fibroblasts (hFBs) populating the graft wall. Using an electrospun composite of polycaprolactone (PCL) and gelatin, we evaluated physical parameters including fibre thickness, fibre alignment, and porosity, that best mimicked the spatial composition and growth of hCAECs and hFBs in native vessels. Upon identifying the optimal scaffold architectures for each cell type, we constructed a custom-designed mandrel that combined these distinct architectures into a single vascular graft during a single electrospinning processing run. When connected to a perfusion bioreactor system, the dual architecture graft spatially oriented hCAECs and hFBs into the graft wall and lumen, respectively, directly from circulation. This biomimetic cell organisation was consistent with positive graft remodelling with significant collagen deposition in the graft wall. These findings demonstrate the influence of architectural cues to direct cell growth within vascular graft templates and the future potential of these approaches to more accurately and efficiency produce blood vessel substitutes in bioreactor systems.
Collapse
Affiliation(s)
- Praveesuda Michael
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Nianji Yang
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Matthew Moore
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Miguel Santos
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Yuen Ting Lam
- The University of Sydney Charles Perkins Centre, John Hopkins Drive, CAMPERDOWN, Sydney, 2006, AUSTRALIA
| | - Annabelle Ward
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Jui Chien Hung
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Richard Tan
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Steven Wise
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| |
Collapse
|
28
|
Chow B, Warkentin B, McEwen M, Huang F, Nanda K, Gamper AM, Menon G. Uncertainties Associated with Clonogenic Assays using a Cs-137 Irradiator and Ir-192 Afterloader: A Comprehensive Compilation for Radiation Researchers. Radiat Res 2022; 198:40-56. [PMID: 35391488 DOI: 10.1667/rade-21-00205.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
Abstract
Clonogenic assays are the gold standard for measuring cell clonogenic survival and enable quantification of a cell line's radiosensitivity through the calculation of the surviving fraction, the ratio of cell clusters (colonies) formed after radiation exposure compared to the number formed without exposure. Such studies regularly utilize Cs-137 irradiators. While uncertainties for specific procedural aspects have been described previously, a comprehensive review has not been completed. We therefore quantified uncertainties associated with clonogenic assays performed using a Cs-137 Shepherd irradiator, and a recently established brachytherapy afterloader in vitro radiation delivery apparatus (BAIRDA), through a series of experiments and a literature review. The clonogenic assay is subject to uncertainties that affect the determination of the surviving fraction (e.g., accuracy of the number of cells seeded, potential effects of hypothermia, and the threshold number of cells for a cluster to be identified as a colony). Furthermore, dose delivery uncertainties related to both the Cs-137 irradiator and BAIRDA were also quantified. The combined standard (k = 1) uncertainty was ± 6.0% in the surviving fraction for the Cs-137 irradiator (±6.3% for BAIRDA), up to ± 1.3% in the dose delivered by the Cs-137 irradiator, and up to ± 2.2% in the dose delivered by BAIRDA. The largest individual uncertainties were associated with the number of cells seeded on a plate (3.4%) and inter-observer variability in counting (4.1%), suggesting that effective reduction of uncertainties in the conduct of the clonogenic assay proper may provide the greatest relief on the uncertainty budget. Finally, measurable impact on experimental findings was assessed by applying this uncertainty to clonogenic assays of SW756 cells using either a Cs-137 irradiator or BAIRDA, introducing a maximum shift in the reported radiobiological parameters a/b and T1/2 of 0.3 Gy and 0.4 h, respectively, while the 95% confidence interval increased by 0.5 Gy and decreased by 0.4 h, respectively. Though the overall impact on radiobiological parameter estimation was small, the individual uncertainties could have a significant influence in other applications of in vitro experiments in radiation biology. Hence, better understanding of the uncertainties associated with both clonogenic assays and the radiation source used can improve the accuracy of experimental analysis and reproducibility of the results.
Collapse
Affiliation(s)
- Braden Chow
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Brad Warkentin
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Malcolm McEwen
- Ionizing Radiation Standards, National Research Council of Canada, Ottawa, Canada
| | - Fleur Huang
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Kareena Nanda
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Armin M Gamper
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Geetha Menon
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
29
|
Shaikh MS, Shahzad Z, Tash EA, Janjua OS, Khan MI, Zafar MS. Human Umbilical Cord Mesenchymal Stem Cells: Current Literature and Role in Periodontal Regeneration. Cells 2022; 11:cells11071168. [PMID: 35406732 PMCID: PMC8997495 DOI: 10.3390/cells11071168] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Periodontal disease can cause irreversible damage to tooth-supporting tissues such as the root cementum, periodontal ligament, and alveolar bone, eventually leading to tooth loss. While standard periodontal treatments are usually helpful in reducing disease progression, they cannot repair or replace lost periodontal tissue. Periodontal regeneration has been demonstrated to be beneficial in treating intraosseous and furcation defects to varied degrees. Cell-based treatment for periodontal regeneration will become more efficient and predictable as tissue engineering and progenitor cell biology advance, surpassing the limitations of present therapeutic techniques. Stem cells are undifferentiated cells with the ability to self-renew and differentiate into several cell types when stimulated. Mesenchymal stem cells (MSCs) have been tested for periodontal regeneration in vitro and in humans, with promising results. Human umbilical cord mesenchymal stem cells (UC-MSCs) possess a great regenerative and therapeutic potential. Their added benefits comprise ease of collection, endless source of stem cells, less immunorejection, and affordability. Further, their collection does not include the concerns associated with human embryonic stem cells. The purpose of this review is to address the most recent findings about periodontal regenerative mechanisms, different stem cells accessible for periodontal regeneration, and UC-MSCs and their involvement in periodontal regeneration.
Collapse
Affiliation(s)
- Muhammad Saad Shaikh
- Department of Oral Biology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi 75510, Pakistan;
| | - Zara Shahzad
- Lahore Medical and Dental College, University of Health Sciences, Lahore 53400, Pakistan;
| | - Esraa Abdulgader Tash
- Department of Oral and Clinical Basic Science, College of Dentistry, Taibah University, Al Madinah Al Munawarah 41311, Saudi Arabia;
| | - Omer Sefvan Janjua
- Department of Maxillofacial Surgery, PMC Dental Institute, Faisalabad Medical University, Faisalabad 38000, Pakistan;
| | | | - Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah Al Munawarah 41311, Saudi Arabia
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
- Correspondence: ; Tel.: +966-507544691
| |
Collapse
|
30
|
Vascular Remodeling of Clinically Used Patches and Decellularized Pericardial Matrices Recellularized with Autologous or Allogeneic Cells in a Porcine Carotid Artery Model. Int J Mol Sci 2022; 23:ijms23063310. [PMID: 35328732 PMCID: PMC8954945 DOI: 10.3390/ijms23063310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Cardiovascular surgery is confronted by a lack of suitable materials for patch repair. Acellular animal tissues serve as an abundant source of promising biomaterials. The aim of our study was to explore the bio-integration of decellularized or recellularized pericardial matrices in vivo. Methods: Porcine (allograft) and ovine (heterograft, xenograft) pericardia were decellularized using 1% sodium dodecyl sulfate ((1) Allo-decel and (2) Xeno-decel). We used two cell types for pressure-stimulated recellularization in a bioreactor: autologous adipose tissue-derived stromal cells (ASCs) isolated from subcutaneous fat of pigs ((3) Allo-ASC and (4) Xeno-ASC) and allogeneic Wharton’s jelly mesenchymal stem cells (WJCs) ((5) Allo-WJC and (6) Xeno-WJC). These six experimental patches were implanted in porcine carotid arteries for one month. For comparison, we also implanted six types of control patches, namely, arterial or venous autografts, expanded polytetrafluoroethylene (ePTFE Propaten® Gore®), polyethylene terephthalate (PET Vascutek®), chemically stabilized bovine pericardium (XenoSure®), and detoxified porcine pericardium (BioIntegral® NoReact®). The grafts were evaluated through the use of flowmetry, angiography, and histological examination. Results: All grafts were well-integrated and patent with no signs of thrombosis, stenosis, or aneurysm. A histological analysis revealed that the arterial autograft resembled a native artery. All other control and experimental patches developed neo-adventitial inflammation (NAI) and neo-intimal hyperplasia (NIH), and the endothelial lining was present. NAI and NIH were most prominent on XenoSure® and Xeno-decel and least prominent on NoReact®. In xenografts, the degree of NIH developed in the following order: Xeno-decel > Xeno-ASC > Xeno-WJC. NAI and patch resorption increased in Allo-ASC and Xeno-ASC and decreased in Allo-WJC and Xeno-WJC. Conclusions: In our setting, pre-implant seeding with ASC or WJC had a modest impact on vascular patch remodeling. However, ASC increased the neo-adventitial inflammatory reaction and patch resorption, suggesting accelerated remodeling. WJC mitigated this response, as well as neo-intimal hyperplasia on xenografts, suggesting immunomodulatory properties.
Collapse
|
31
|
Davari N, Bakhtiary N, Khajehmohammadi M, Sarkari S, Tolabi H, Ghorbani F, Ghalandari B. Protein-Based Hydrogels: Promising Materials for Tissue Engineering. Polymers (Basel) 2022; 14:986. [PMID: 35267809 PMCID: PMC8914701 DOI: 10.3390/polym14050986] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
The successful design of a hydrogel for tissue engineering requires a profound understanding of its constituents' structural and molecular properties, as well as the proper selection of components. If the engineered processes are in line with the procedures that natural materials undergo to achieve the best network structure necessary for the formation of the hydrogel with desired properties, the failure rate of tissue engineering projects will be significantly reduced. In this review, we examine the behavior of proteins as an essential and effective component of hydrogels, and describe the factors that can enhance the protein-based hydrogels' structure. Furthermore, we outline the fabrication route of protein-based hydrogels from protein microstructure and the selection of appropriate materials according to recent research to growth factors, crucial members of the protein family, and their delivery approaches. Finally, the unmet needs and current challenges in developing the ideal biomaterials for protein-based hydrogels are discussed, and emerging strategies in this area are highlighted.
Collapse
Affiliation(s)
- Niyousha Davari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 143951561, Iran;
| | - Negar Bakhtiary
- Burn Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran 14115114, Iran
| | - Mehran Khajehmohammadi
- Department of Mechanical Engineering, Faculty of Engineering, Yazd University, Yazd 8174848351, Iran;
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd 8916877391, Iran
| | - Soulmaz Sarkari
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran;
| | - Hamidreza Tolabi
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran 158754413, Iran;
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 158754413, Iran
| | - Farnaz Ghorbani
- Institute of Biomaterials, Department of Material Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
32
|
Current Progress in Vascular Engineering and Its Clinical Applications. Cells 2022; 11:cells11030493. [PMID: 35159302 PMCID: PMC8834640 DOI: 10.3390/cells11030493] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Coronary heart disease (CHD) is caused by narrowing or blockage of coronary arteries due to atherosclerosis. Coronary artery bypass grafting (CABG) is widely used for the treatment of severe CHD cases. Although autologous vessels are a preferred choice, healthy autologous vessels are not always available; hence there is a demand for tissue engineered vascular grafts (TEVGs) to be used as alternatives. However, producing clinical grade implantable TEVGs that could healthily survive in the host with long-term patency is still a great challenge. There are additional difficulties in producing small diameter (<6 mm) vascular conduits. As a result, there have not been TEVGs that are commercially available. Properties of vascular scaffolds such as tensile strength, thrombogenicity and immunogenicity are key factors that determine the biocompatibility of TEVGs. The source of vascular cells employed to produce TEVGs is a limiting factor for large-scale productions. Advanced technologies including the combined use of natural and biodegradable synthetic materials for scaffolds in conjunction with the use of mesenchyme stem cells or induced pluripotent stem cells (iPSCs) provide promising solutions for vascular tissue engineering. The aim of this review is to provide an update on various aspects in this field and the current status of TEVG clinical applications.
Collapse
|
33
|
Semitela Â, Ramalho G, Capitão A, Sousa C, Mendes AF, Aap Marques P, Completo A. Bio-electrospraying assessment toward in situ chondrocyte-laden electrospun scaffold fabrication. J Tissue Eng 2022; 13:20417314211069342. [PMID: 35024136 PMCID: PMC8743920 DOI: 10.1177/20417314211069342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 01/05/2023] Open
Abstract
Electrospinning has been widely used to fabricate fibrous scaffolds for cartilage tissue engineering, but their small pores severely restrict cell infiltration, resulting in an uneven distribution of cells across the scaffold, particularly in three-dimensional designs. If bio-electrospraying is applied, direct chondrocyte incorporation into the fibers during electrospinning may be a solution. However, before this approach can be effectively employed, it is critical to identify whether chondrocytes are adversely affected. Several electrospraying operating settings were tested to determine their effect on the survival and function of an immortalized human chondrocyte cell line. These chondrocytes survived through an electric field formed by low needle-to-collector distances and low voltage. No differences in chondrocyte viability, morphology, gene expression, or proliferation were found. Preliminary data of the combination of electrospraying and polymer electrospinning disclosed that chondrocyte integration was feasible using an alternated approach. The overall increase in chondrocyte viability over time indicated that the embedded cells retained their proliferative capacity. Besides the cell line, primary chondrocytes were also electrosprayed under the previously optimized operational conditions, revealing the higher sensitivity degree of these cells. Still, their post-electrosprayed viability remained considerably high. The data reported here further suggest that bio-electrospraying under the optimal operational conditions might be a promising alternative to the existent cell seeding techniques, promoting not only cells safe delivery to the scaffold, but also the development of cellularized cartilage tissue constructs.
Collapse
Affiliation(s)
- Ângela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Gonçalo Ramalho
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Ana Capitão
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Cátia Sousa
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Alexandrina F Mendes
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Paula Aap Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
34
|
Bae JY, Choi SJ, Kim JK. Comparison of mesenchymal stem cell attachment efficiency in acellular neural graft for peripheral nerve regeneration. J Plast Reconstr Aesthet Surg 2021; 75:1674-1681. [PMID: 34955403 DOI: 10.1016/j.bjps.2021.11.098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 02/06/2023]
Abstract
Decellularized nerve allograft is an alternative to autologous nerve graft for nerve defects but has shown inferior clinical outcomes. Mesenchymal stem cells can play a key role in improving nerve regeneration of decellularized nerve allografts. The purpose of this study was to compare different mesenchymal stem cell seeding methods and to find the most efficient way to attach cells to nerve grafts for peripheral nerve regeneration. Wharton's jelly mesenchymal stem cells were collected from human umbilical cords and were seeded in the acellular nerve graft in five different ways as follows: PBS injection, fibrin glue drop, Matrigel drop, bioreactor, and Matrigel injection. A 6-mm sciatic nerve defect of Sprague-Dawley rats was bridged using mesenchymal stem cells-laden acellular nerve grafts according to the five seeding methods. Two days after implantation, the nerve tissue was biopsied and analyzed by the immunofluorescence staining of nuclei. The number of Wharton's jelly mesenchymal stem cells (+ h Nuclei) was counted in the inside, outside, and the total area of the graft sections under 200X magnification. The highest efficiency of mesenchymal stem cell attachment inside the graft and the highest total number of attached mesenchymal stem cells was observed in the group using Matrigel injection (p < 0.0001). This study showed mesenchymal stem cells can be more effectively attached to decellularized nerve graft using the injection method with Matrigel than other static or dynamic seeding methods in vivo.
Collapse
Affiliation(s)
- Joo-Yul Bae
- Department of Orthopedic Surgery, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung-si, Korea
| | | | - Jae Kwang Kim
- Department of Orthopedic Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
35
|
Durán-Rey D, Crisóstomo V, Sánchez-Margallo JA, Sánchez-Margallo FM. Systematic Review of Tissue-Engineered Vascular Grafts. Front Bioeng Biotechnol 2021; 9:771400. [PMID: 34805124 PMCID: PMC8595218 DOI: 10.3389/fbioe.2021.771400] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/18/2021] [Indexed: 01/01/2023] Open
Abstract
Pathologies related to the cardiovascular system are the leading causes of death worldwide. One of the main treatments is conventional surgery with autologous transplants. Although donor grafts are often unavailable, tissue-engineered vascular grafts (TEVGs) show promise for clinical treatments. A systematic review of the recent scientific literature was performed using PubMed (Medline) and Web of Science databases to provide an overview of the state-of-the-art in TEVG development. The use of TEVG in human patients remains quite restricted owing to the presence of vascular stenosis, existence of thrombi, and poor graft patency. A total of 92 original articles involving human patients and animal models were analyzed. A meta-analysis of the influence of the vascular graft diameter on the occurrence of thrombosis and graft patency was performed for the different models analyzed. Although there is no ideal animal model for TEVG research, the murine model is the most extensively used. Hybrid grafting, electrospinning, and cell seeding are currently the most promising technologies. The results showed that there is a tendency for thrombosis and non-patency in small-diameter grafts. TEVGs are under constant development, and research is oriented towards the search for safe devices.
Collapse
Affiliation(s)
- David Durán-Rey
- Laparoscopy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Verónica Crisóstomo
- Cardiovascular Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.,Centro de Investigacion Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan A Sánchez-Margallo
- Bioengineering and Health Technologies Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Francisco M Sánchez-Margallo
- Centro de Investigacion Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Scientific Direction, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| |
Collapse
|
36
|
Double w/o/w self-nano emulsifying drug delivery system of imatinib mesylate for colon cancer treatment. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Devillard CD, Marquette CA. Vascular Tissue Engineering: Challenges and Requirements for an Ideal Large Scale Blood Vessel. Front Bioeng Biotechnol 2021; 9:721843. [PMID: 34671597 PMCID: PMC8522984 DOI: 10.3389/fbioe.2021.721843] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/20/2021] [Indexed: 01/05/2023] Open
Abstract
Since the emergence of regenerative medicine and tissue engineering more than half a century ago, one obstacle has persisted: the in vitro creation of large-scale vascular tissue (>1 cm3) to meet the clinical needs of viable tissue grafts but also for biological research applications. Considerable advancements in biofabrication have been made since Weinberg and Bell, in 1986, created the first blood vessel from collagen, endothelial cells, smooth muscle cells and fibroblasts. The synergistic combination of advances in fabrication methods, availability of cell source, biomaterials formulation and vascular tissue development, promises new strategies for the creation of autologous blood vessels, recapitulating biological functions, structural functions, but also the mechanical functions of a native blood vessel. In this review, the main technological advancements in bio-fabrication are discussed with a particular highlights on 3D bioprinting technologies. The choice of the main biomaterials and cell sources, the use of dynamic maturation systems such as bioreactors and the associated clinical trials will be detailed. The remaining challenges in this complex engineering field will finally be discussed.
Collapse
Affiliation(s)
- Chloé D Devillard
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Christophe A Marquette
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| |
Collapse
|
38
|
Munteanu C, Mireşan V, Răducu C, Ihuţ A, Uiuiu P, Pop D, Neacşu A, Cenariu M, Groza I. Can Cultured Meat Be an Alternative to Farm Animal Production for a Sustainable and Healthier Lifestyle? Front Nutr 2021; 8:749298. [PMID: 34671633 PMCID: PMC8522976 DOI: 10.3389/fnut.2021.749298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
Producing animal proteins requires large areas of agricultural land and is a major source of greenhouse gases. Cellular agriculture, especially cultured meat, could be a potential alternative for the environment and human health. It enables meat and other agricultural products to be grown from cells in a bioreactor without being taken from farm animals. This paper aims at an interdisciplinary review of literature focusing on potential benefits and risks associated with cultured meat. To achieve this goal, several international databases and governmental projects were thoroughly analyzed using keywords and phrases with specialty terms. This is a growing scientific domain, which has generated a series of debates regarding its potential effects. On the one hand the potential of beneficial effects is the reduction of agricultural land usage, pollution and the improvement of human health. Other authors question if cultured meat could be a sustainable alternative for reducing gas emissions. Interestingly, the energy used for cultured meat could be higher, due to the replacement of some biological functions, by technological processes. For potential effects to turn into results, a realistic understanding of the technology involved and more experimental studies are required.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Vioara Mireşan
- Department of Fundamental Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Camelia Răducu
- Department of Technological Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Andrada Ihuţ
- Department of Technological Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Paul Uiuiu
- Department of Fundamental Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Daria Pop
- Clinic of Obstetrics and Gynecology II "Dominic Stanca, " University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Cluj-Napoca, Romania
| | - Alexandra Neacşu
- Department of Chemical Engineering, Babeş-Bolyai University, Cluj-Napoca, Romania
| | - Mihai Cenariu
- Department of Animal Reproduction and Reproductive Pathology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Ioan Groza
- Department of Animal Reproduction and Reproductive Pathology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
39
|
Abstract
Decellularization technology is a process that uses different methods such as physical, chemical or enzymatic methods in order to eliminate cellular remnants from original tissues or organs while minimizing any adverse effect on the structural properties, biological activity, and mechanical integrity of the remaining ECM. Regenerative medicine uses the most promising therapies to replace or regenerate tissues and organs in human, restore or establish normal functions lost due to disease or injury. By the combination between new biomaterials and cells, one of the goals of regenerative medicine is to create autologous grafts for transplantation therapies in the future.Various decellularization methods have been developed include chemical treatment, biological treatment and physical treatment. The aim of this chapter is to evaluate the decellularization method and all available materials that preserves the matrix without structural disruption.
Collapse
|
40
|
Kim SW, Kim YY, Kim H, Ku SY. Recent Advancements in Engineered Biomaterials for the Regeneration of Female Reproductive Organs. Reprod Sci 2021; 28:1612-1625. [PMID: 33797052 DOI: 10.1007/s43032-021-00553-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/17/2021] [Indexed: 12/14/2022]
Abstract
Various gynecologic diseases and chemoradiation or surgery for the management of gynecologic malignancies may damage the uterus and ovaries, leading to clinical problems such as infertility or early menopause. Embryo or oocyte cryopreservation-the standard method for fertility preservation-is not a feasible option for patients who require urgent treatment because the procedure requires ovarian stimulation for at least several days. Hormone replacement therapy (HRT) for patients diagnosed with premature menopause is contraindicated for patients with estrogen-dependent tumors or a history of thrombosis. Furthermore, these methods cannot restore the function of the uterus and ovaries. Although autologous transplantation of cryopreserved ovarian tissue is being attempted, it may re-introduce malignant cells after cancer treatment. With the recent development in regenerative medicine, research on engineered biomaterials for the restoration of female reproductive organs is being actively conducted. The use of engineered biomaterials is a promising option in the field of reproductive medicine because it can overcome the limitations of current therapies. Here, we review the ideal properties of biomaterials for reproductive tissue engineering and the recent advancements in engineered biomaterials for the regeneration of female reproductive organs.
Collapse
Affiliation(s)
- Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, 03080, South Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, 03080, South Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea.
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, 03080, South Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, 2024 E. Monument St, Baltimore, MD, 21205, USA.
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, 03080, South Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
41
|
Chitosan scaffolds with enhanced mechanical strength and elastic response by combination of freeze gelation, photo-crosslinking and freeze-drying. Carbohydr Polym 2021; 267:118156. [PMID: 34119130 DOI: 10.1016/j.carbpol.2021.118156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/01/2021] [Accepted: 04/16/2021] [Indexed: 01/17/2023]
Abstract
In this study, a new scaffold fabrication method based on the combination of a series of stabilization processes was set up to obtain chitosan scaffolds with improved mechanical properties for regeneration of load-bearing tissues. Specifically, thermally induced phase separation (TIPS) of chitosan solutions was used to obtain an open structure which was then stabilized by freeze-gelation and photo cross-linking. Freeze-gelation combined with freeze-drying permitted to obtain a porous structure with a 95 μm-mean pore size suitable for osteoblast cells' housing. Photo-crosslinking improved by ca. three times the scaffold compressive modulus, passing from 0,8 MPa of the uncrosslinked scaffolds to 2,2 MPa of the crosslinked one. Hydrated crosslinked scaffolds showed a good elastic response, with an 80% elastic recovery for at least 5 consecutive compressive cycles. The herein reported method has the advantage to not require the use of potentially toxic cross-linking agents and may be extended to other soft materials.
Collapse
|
42
|
Gorji A, Toh PJY, Ong HT, Toh YC, Toyama Y, Kanchanawong P. Enhancement of Endothelialization by Topographical Features Is Mediated by PTP1B-Dependent Endothelial Adherens Junctions Remodeling. ACS Biomater Sci Eng 2021; 7:2661-2675. [PMID: 33942605 DOI: 10.1021/acsbiomaterials.1c00251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endothelial Cells (ECs) form cohesive cellular lining of the vasculature and play essential roles in both developmental processes and pathological conditions. Collective migration and proliferation of endothelial cells (ECs) are key processes underlying endothelialization of vessels as well as vascular graft, but the complex interplay of mechanical and biochemical signals regulating these processes are still not fully elucidated. While surface topography and biochemical modifications have been used to enhance endothelialization in vitro, thus far such single-modality modifications have met with limited success. As combination therapy that utilizes multiple modalities has shown improvement in addressing various intractable and complex biomedical conditions, here, we explore a combined strategy that utilizes topographical features in conjunction with pharmacological perturbations. We characterized EC behaviors in response to micrometer-scale grating topography in concert with pharmacological perturbations of endothelial adherens junctions (EAJ) regulators. We found that the protein tyrosine phosphatase, PTP1B, serves as a potent regulator of EAJ stability, with PTP1B inhibition synergizing with grating topographies to modulate EAJ rearrangement, thereby augmenting global EC monolayer sheet orientation, proliferation, connectivity, and collective cell migration. Our data delineates the crosstalk between cell-ECM topography sensing and cell-cell junction integrity maintenance and suggests that the combined use of grating topography and PTP1B inhibitor could be a promising strategy for promoting collective EC migration and proliferation.
Collapse
Affiliation(s)
- Azita Gorji
- Mechanobiology Institute, National University of Singapore, 117411 Republic of Singapore.,Institut Curie, Laboratoire Physico Chimie Curie, Institut Pierre-Gilles de Gennes, CNRS UMR168, Paris 75005, France
| | - Pearlyn Jia Ying Toh
- Mechanobiology Institute, National University of Singapore, 117411 Republic of Singapore
| | - Hui Ting Ong
- Mechanobiology Institute, National University of Singapore, 117411 Republic of Singapore
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 117583 Republic of Singapore.,Institute for Health Innovation and Technology, National University of Singapore, 117599 Republic of Singapore.,The N.1 Institute for Health, National University of Singapore, 117456, Republic of Singapore.,NUS Tissue Engineering Programme, National University of Singapore, 117456, Republic of Singapore
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, 117411 Republic of Singapore.,Department of Biological Sciences, National University of Singapore, 117558, Republic of Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, 117411 Republic of Singapore.,Department of Biomedical Engineering, National University of Singapore, 117583 Republic of Singapore
| |
Collapse
|
43
|
Nedunchezian S, Banerjee P, Lee CY, Lee SS, Lin CW, Wu CW, Wu SC, Chang JK, Wang CK. Generating adipose stem cell-laden hyaluronic acid-based scaffolds using 3D bioprinting via the double crosslinked strategy for chondrogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112072. [PMID: 33947564 DOI: 10.1016/j.msec.2021.112072] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/26/2021] [Accepted: 03/13/2021] [Indexed: 12/20/2022]
Abstract
Bioprinting of most cell-laden hydrogel scaffolds with the required structural integrity, mechanical modulus, cell adhesion, cell compatibility, and chondrogenic differentiation are still significant issues that affect the application of bioinks in cartilage tissue engineering. This study focuses on constructing printable bioinks by combining adipose-derived stem cells (ADSCs), hyaluronic acid (HA)-based hydrogels and analyzing their ability to induce chondrogenesis using 3D bioprinting technology. First, biotinylated hyaluronic acid was synthesized via an adipic acid dihydrazide (ADH) linker with amide bond formation to form HA-biotin (HAB). Both HAB and the as-received streptavidin were mixed to form a partially cross-linked HA-biotin-streptavidin (HBS) hydrogel through noncovalent bonding. After that, the partially cross-linked HBS hydrogel was mixed with sodium alginate and subsequently printed to form the HBSA hydrogel 3D scaffolds using a bioprinter. Finally, the 3D scaffolds of the HBSA (HBS + alginate) hydrogel were submerged into CaCl2 solution to achieve a stable 3D HBSAC (HBSA + Ca2+) hydrogel scaffold through ion transfer crosslinking. The physical-chemical characteristics of the hybrid bioink compositions have been evaluated to determine the desired 3D bioprinting structure. Cytotoxicity and chondrogenic differentiation were also assessed to confirm that the double cross-linked HBSAC hydrogel scaffold was useful for chondrogenic formation. The results showed that partially crosslinking the biotinylated HA-based hydrogel with streptavidin has a significant effect on printability and structural integrity. Morphological analysis of a suitable 3D printed HBSAC hydrogel scaffold showed visible pores with the desired shape and geometry. We have concluded that the HBSAC hydrogel possesses a favorable biocompatibility profile. The HBSAC hydrogel can also secrete significantly higher amounts of chondrogenic marker genes at day 5 and sulfated glycosaminoglycans (sGAGs) from days 7 to 14 compared to the HA hydrogel, as determined via quantitative real-time PCR assay and Alcian blue staining and the DMMB assay.
Collapse
Affiliation(s)
- Swathi Nedunchezian
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Parikshit Banerjee
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Yun Lee
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Ph.D Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Su-Shin Lee
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Che-Wei Lin
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Che-Wei Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shun-Cheng Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Kuang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Ph.D Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
44
|
Evaluation of the regenerative potential of decellularized skeletal muscle seeded with mesenchymal stromal cells in critical-sized bone defect of rat models. Saudi Dent J 2021; 33:248-255. [PMID: 34194187 PMCID: PMC8236553 DOI: 10.1016/j.sdentj.2021.03.006] [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: 02/10/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/20/2022] Open
Abstract
Background The morbidities and complications reported in the reconstruction of large bony defects have inspired progression in the field of bioengineering, with a recent breakthrough for the use of decellularized skeletal muscle grafts (DSMG). Aim To assess the osteogenic potentials of seeded DSMG in vitro and to investigate bone regeneration in critical size defect in vivo. Materials and Methods Assessment of cell viability and characterization was carried out on seeded DSMG for different intervals in vitro. For in vivo experiments, histological analysis was performed for rat cranial defects for the following groups: (A) non-treated DSMG and (B) seeded DSMG after a period of 8 weeks. Results The in vitro experiment demonstrated the lack of cytotoxicity and inert properties of seeded DSMG; these facilitated the osteogenic differentiation and significant gene expressions, particularly of COL1A1, RUNX2, and OPN (1.9174 ± 0.11673, 1.1806 ± 0.02383, and 1.1802 ± 0.00775, respectively). In the in vivo experiment, superior results were detected in the seeded DSMG group which showed highly vascularized and cellular dense connective tissue with deposited bone matrix and multiple scattered islets of newly formed bone. Conclusion Our results demonstrated the promising aspects of DSMG; however, there is a lack of studies to support further implications.
Collapse
|
45
|
Asadi M, Khalili M, Lotfi H, Vaghefi Moghaddam S, Zarghami N, André H, Alizadeh E. Liver bioengineering: Recent trends/advances in decellularization and cell sheet technologies towards translation into the clinic. Life Sci 2021; 276:119373. [PMID: 33744324 DOI: 10.1016/j.lfs.2021.119373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Development of novel technologies provides the best tissue constructs engineering and maximizes their therapeutic effects in regenerative therapy, especially for liver dysfunctions. Among the currently investigated approaches of tissue engineering, scaffold-based and scaffold-free tissues are widely suggested for liver regeneration. Analogs of liver acellular extracellular matrix (ECM) are utilized in native scaffolds to increase the self-repair and healing ability of organs. Native ECM analog could improve liver repairing through providing the supportive framework for cells and signaling molecules, exerting normal biomechanical, biochemical, and physiological signal complexes. Recently, innovative cell sheet technology is introduced as an alternative for conventional tissue engineering with the advantage of fewer scaffold restrictions and cell culture on a Thermo-Responsive Polymer Surface. These sheets release the layered cells through a temperature-controlled procedure without enzymatic digestion, while preserving the cell-ECM contacts and adhesive molecules on cell-cell junctions. In addition, several novelties have been introduced into the cell sheet and decellularization technologies to aid cell growth, instruct differentiation/angiogenesis, and promote cell migration. In this review, recent trends, advancements, and issues linked to translation into clinical practice are dissected and compared regarding the decellularization and cell sheet technologies for liver tissue engineering.
Collapse
Affiliation(s)
- Maryam Asadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Khalili
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helder André
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institute, 11282 Stockholm, Sweden
| | - Effat Alizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
46
|
Muthukrishnan L. Imminent antimicrobial bioink deploying cellulose, alginate, EPS and synthetic polymers for 3D bioprinting of tissue constructs. Carbohydr Polym 2021; 260:117774. [PMID: 33712131 DOI: 10.1016/j.carbpol.2021.117774] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/16/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022]
Abstract
3D printing, one of its kinds has been a recent technological trend to fabricate complex and patterned biomaterial with controlled precision. With the conventional kick-start of printing metals and plastics, advancements in printing viable cells, polysaccharides or microbes themselves have been achieved. The additive antimicrobial properties in bioinks sourced from organic and inorganic materials have profound implications in tissue engineering. Cellulose, alginate, exopolysaccharides, ceramics and synthetic polymers are integrated as a viable component in inks and used for bio-printing. To date, bacterial infection and immunogenicity pose a potential health risk during a tissue implant or bone substitution. In order to mitigate microbial infection, antimicrobial bioinks with significant antimicrobial potential have been the much sought after strategies. This approach could be an effective frontline defense against microbial interference in tissue engineering and biomedical applications. An overview on the antimicrobial potential of polysaccharides as bioinks for 3D bioprinting has been critically reviewed.
Collapse
Affiliation(s)
- Lakshmipathy Muthukrishnan
- Department of Conservative Dentistry & Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Poonamallee High Road, Chennai, Tamil Nadu, 600 077, India.
| |
Collapse
|
47
|
Mainardi VL, Arrigoni C, Bianchi E, Talò G, Delcogliano M, Candrian C, Dubini G, Levi M, Moretti M. Improving cell seeding efficiency through modification of fiber geometry in 3D printed scaffolds. Biofabrication 2021; 13. [PMID: 33578401 DOI: 10.1088/1758-5090/abe5b4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/12/2021] [Indexed: 11/11/2022]
Abstract
Cell seeding on 3D scaffolds is a very delicate step in tissue engineering applications, influencing the outcome of the subsequent culture phase, and determining the results of the entire experiment. Thus, it is crucial to maximize its efficiency. To this purpose, a detailed study of the influence of the geometry of the scaffold fibers on dynamic seeding efficiency is presented. 3D printing technology was used to realize PLA porous scaffolds, formed by fibers with a non-circular cross-sectional geometry, named multilobed to highlight the presence of niches and ridges. An oscillating perfusion bioreactor was used to perform bidirectional dynamic seeding of MG63 cells. The fiber shape influences the fluid dynamic parameters of the flow, affecting values of fluid velocity and wall shear stress. The path followed by cells through the scaffold fibers is also affected and results in a larger number of adhered cells in multilobed scaffolds compared to scaffolds with standard pseudo cylindrical fibers. Geometrical and fluid dynamic features can also have an influence on the morphology of adhered cells. The obtained results suggest that the reciprocal influence of geometrical and fluid dynamic features and their combined effect on cell trajectories should be considered to improve the dynamic seeding efficiency when designing scaffold architecture.
Collapse
Affiliation(s)
- Valerio Luca Mainardi
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Elena Bianchi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, ITALY
| | - Giuseppe Talò
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi, 4, Milano, 20161, ITALY
| | - Marco Delcogliano
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Christian Candrian
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| | - Gabriele Dubini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, ITALY
| | - Marinella Levi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, ITALY
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, Via Tesserete, 46, Lugano, 6900, SWITZERLAND
| |
Collapse
|
48
|
Joshi P, Breaux S, Naro J, Wang Y, Ahmed MSU, Vig K, Auad ML. Synthesis and characterization of photopolymerizable hydrogels based on poly (ethylene glycol) for biomedical applications. J Appl Polym Sci 2021. [DOI: 10.1002/app.50489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Prutha Joshi
- Department of Chemical Engineering Auburn University Auburn Alabama USA
- Center for Polymers and Advanced Composites Auburn University Auburn Alabama USA
| | - Steven Breaux
- Department of Chemical Engineering Auburn University Auburn Alabama USA
- Center for Polymers and Advanced Composites Auburn University Auburn Alabama USA
| | - Joseph Naro
- Department of Chemical Engineering Auburn University Auburn Alabama USA
- Center for Polymers and Advanced Composites Auburn University Auburn Alabama USA
| | - Yuyang Wang
- Department of Chemical Engineering Auburn University Auburn Alabama USA
- Center for Polymers and Advanced Composites Auburn University Auburn Alabama USA
| | | | - Komal Vig
- Department of Biological Sciences Alabama State University Montgomery Alabama USA
| | - Maria L. Auad
- Department of Chemical Engineering Auburn University Auburn Alabama USA
- Center for Polymers and Advanced Composites Auburn University Auburn Alabama USA
| |
Collapse
|
49
|
Nizami MZI, Nishina Y. Recent Advances in Stem Cells for Dental Tissue Engineering. ENGINEERING MATERIALS FOR STEM CELL REGENERATION 2021:281-324. [DOI: 10.1007/978-981-16-4420-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
50
|
Mirhaidari GJ, Barker JC, Zbinden JC, Santantonio BM, Chang YC, Best CA, Reinhardt JW, Blum KM, Yi T, Breuer CK. Tissue Engineered Vascular Graft Recipient Interleukin 10 Status Is Critical for Preventing Thrombosis. Adv Healthc Mater 2020; 9:e2001094. [PMID: 33073543 PMCID: PMC7936649 DOI: 10.1002/adhm.202001094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Indexed: 01/07/2023]
Abstract
Tissue engineered vascular grafts (TEVGs) are a promising technology, but are hindered by occlusion. Seeding with bone-marrow derived mononuclear cells (BM-MNCs) mitigates occlusion, yet the precise mechanism remains unclear. Seeded cells disappear quickly and potentially mediate an anti-inflammatory effect through paracrine signaling. Here, a series of reciprocal genetic TEVG implantations plus recombinant protein treatment is reported to investigate what role interleukin-10, an anti-inflammatory cytokine, plays from both host and seeded cells. TEVGs seeded with BM-MNCs from wild-type and IL-10 KO mice, plus unseeded grafts, are implanted into wild-type and IL-10 KO mice. Wild-type mice with unseeded grafts also receive recombinant IL-10. Serial ultrasound evaluates occlusion and TEVGs are harvested at 14 d for immunohistochemical analysis. TEVGs in IL-10 KO mice have significantly higher occlusion incidence compared to wild-type mice attributed to acute (<3 d) thrombosis. Cell seeding rescues TEVGs in IL-10 KO mice comparable to wild-type patency. IL-10 from the host and seeded cells do not significantly influence graft inflammation and macrophage phenotype, yet IL-10 treatment shows interesting biologic effects including decreasing cell proliferation and increasing M2 macrophage polarization. IL-10 from the host is critical for preventing TEVG thrombosis and seeded BM-MNCs exert a significant anti-thrombotic effect in IL-10 KO mice.
Collapse
Affiliation(s)
- Gabriel J.M. Mirhaidari
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Jenny C. Barker
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Jacob C. Zbinden
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Brevan M. Santantonio
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Yu-Chun Chang
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Cameron A. Best
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - James W. Reinhardt
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Kevin M. Blum
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Tai Yi
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Christopher K. Breuer
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| |
Collapse
|