1
|
Getova VE, Pinheiro-Machado E, Harmsen MC, Burgess JK, Smink AM. The role of extracellular matrix hydrogels and adipose-derived stromal cells in soft tissue vascularization - A systematic review. BIOMATERIALS ADVANCES 2024; 164:213986. [PMID: 39151272 DOI: 10.1016/j.bioadv.2024.213986] [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: 11/27/2023] [Revised: 07/12/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
Decellularized extracellular matrix (dECM) hydrogels loaded with adipose-derived stromal cells (ASC) or their conditioned medium (ASC CM) present a promising and versatile treatment approach for tissue vascularization and regeneration. These hydrogels are easy to produce, store, personalize, manipulate, and deliver to the target tissue. This literature review aimed to investigate the applications of dECM hydrogels with ASC or ASC CM for in vivo tissue vascularization. Fourteen experimental studies have been reviewed using vessel density as the primary outcome parameter for in vivo vascularization. The studies consistently reported an increased efficacy in augmenting angiogenesis by the ASC or ASC CM-loaded hydrogels compared to untreated controls. However, this systematic review shows the need to standardize procedures and characterization, particularly of the final administered product(s). The findings from these experimental studies highlight the potential of dECM hydrogel with ASC or ASC CM in novel tissue regeneration and regenerative medicine applications.
Collapse
Affiliation(s)
- Vasilena E Getova
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands
| | - Erika Pinheiro-Machado
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Martin C Harmsen
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Alexandra M Smink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| |
Collapse
|
2
|
Kanniyappan H, Sundaram MK, Ravikumar A, Chakraborty S, Gnanamani A, Mani U, Kumar N, Muthuvijayan V. Enhancing bone repair through improved angiogenesis and osteogenesis using mesoporous silica nanoparticle-loaded Konjac glucomannan-based interpenetrating network scaffolds. Int J Biol Macromol 2024; 279:135182. [PMID: 39216566 DOI: 10.1016/j.ijbiomac.2024.135182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/17/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
We have fabricated and characterized novel bioactive nanocomposite interpenetrating polymer network (IPN) scaffolds to treat bone defects by loading mesoporous silica nanoparticles (MSNs) into blends of Konjac glucomannan, polyvinyl alcohol, and polycaprolactone. By loading MSNs, we developed a porous nanocomposite scaffold with mechanical strengths comparable to cancellous bone. In vitro cell culture studies proved the cytocompatibility of the nanocomposite scaffolds. RT-PCR studies confirmed that these scaffolds significantly upregulated major osteogenic markers. The in vivo chick chorioallantoic membrane (CAM) assay confirmed the proangiogenic activity of the nanocomposite IPN scaffolds. In vivo studies were performed using Wistar rats to evaluate the scaffolds' compatibility, osteogenic activity, and proangiogenic properties. Liver and renal function tests confirmed that these scaffolds were nontoxic. X-ray and μ-CT results show that the bone defects treated with the nanocomposite scaffolds healed at a much faster rate compared to the untreated control and those treated with IPN scaffolds. H&E and Masson's trichrome staining showed angiogenesis near the newly formed bone and the presence of early-stage connective tissues, fibroblasts, and osteoblasts in the defect region at 8 weeks after surgery. Hence, these advantageous physicochemical and biological properties confirm that the nanocomposite IPN scaffolds are ideal for treating bone defects.
Collapse
Affiliation(s)
- Hemalatha Kanniyappan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Manoj Kumar Sundaram
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Akhil Ravikumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Sudip Chakraborty
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| | - A Gnanamani
- Microbiology Lab, CSIR-Central Leather Research Institute, Chennai 600020, India
| | - U Mani
- Animal House, CSIR-Central Leather Research Institute, Chennai 600020, India
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Vignesh Muthuvijayan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
3
|
Barrett P, Louie KW, Dupont JB, Mack DL, Maves L. Uncovering the Embryonic Origins of Duchenne Muscular Dystrophy. WIREs Mech Dis 2024:e1653. [PMID: 39444092 DOI: 10.1002/wsbm.1653] [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: 07/05/2024] [Revised: 09/11/2024] [Accepted: 09/28/2024] [Indexed: 10/25/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a severe degenerative muscle disease caused by mutations in the DMD gene, which encodes dystrophin. Despite its initial description in the late 19th century by French neurologist Guillaume Duchenne de Boulogne, and identification of causal DMD genetic mutations in the 1980s, therapeutics remain challenging. The current standard of care is corticosteroid treatment, which delays the progression of muscle dysfunction but is associated with significant adverse effects. Emerging therapeutic approaches, including AAV-mediated gene transfer, CRISPR gene editing, and small molecule interventions, are under development but face considerable obstacles. Although DMD is viewed as a progressive muscle disease, muscle damage and abnormal molecular signatures are already evident during fetal myogenesis. This early onset of pathology suggests that the limited success of current therapies may partly be due to their administration after aberrant embryonic myogenesis has occurred in the absence of dystrophin. Consequently, identifying optimal therapeutic strategies and intervention windows for DMD may depend on a better understanding of the earliest DMD disease mechanisms. As newer techniques are applied, the field is gaining increasingly detailed insights into the early muscle developmental abnormalities in DMD. A comprehensive understanding of the initial events in DMD pathogenesis and progression will facilitate the generation and testing of effective therapeutic interventions.
Collapse
Affiliation(s)
- Philip Barrett
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA
| | - Ke'ale W Louie
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | | | - David L Mack
- Departments of Rehabilitation Medicine, Bioengineering and Neurobiology & Biophysics, Institute for Stem Cell and Regenerative Medicine, University of Washington Medicine, Seattle, Washington, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Liu JD, VanTreeck KE, Marston WA, Papadopoulou V, Rowe SE. Ultrasound-Mediated Antibiotic Delivery to In Vivo Biofilm Infections: A Review. Chembiochem 2024; 25:e202400181. [PMID: 38924307 PMCID: PMC11483220 DOI: 10.1002/cbic.202400181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Bacterial biofilms are a significant concern in various medical contexts due to their resilience to our immune system as well as antibiotic therapy. Biofilms often require surgical removal and frequently lead to recurrent or chronic infections. Therefore, there is an urgent need for improved strategies to treat biofilm infections. Ultrasound-mediated drug delivery is a technique that combines ultrasound application, often with the administration of acoustically-active agents, to enhance drug delivery to specific target tissues or cells within the body. This method involves using ultrasound waves to assist in the transportation or activation of medications, improving their penetration, distribution, and efficacy at the desired site. The advantages of ultrasound-mediated drug delivery include targeted and localized delivery, reduced systemic side effects, and improved efficacy of the drug at lower doses. This review scrutinizes recent advances in the application of ultrasound-mediated drug delivery for treating biofilm infections, focusing on in vivo studies. We examine the strengths and limitations of this technology in the context of wound infections, device-associated infections, lung infections and abscesses, and discuss current gaps in knowledge and clinical translation considerations.
Collapse
Affiliation(s)
- Jamie D. Liu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kelly E. VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William A. Marston
- Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Department of Radiology, The University of North Carolina at Chapel Hill, NC, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
5
|
Hînganu D, Hînganu MV, Tamaș C, Costan VV, Hristian L, Negru D, Calistru AE, Cucu RP, Lozneanu L. Vascular Perspectives of the Midfacial Superficial Musculoaponeurotic System. Diagnostics (Basel) 2024; 14:2294. [PMID: 39451617 PMCID: PMC11507235 DOI: 10.3390/diagnostics14202294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
Objectives: Presently, data on the vascularization of the superficial musculoaponeurotic system of the face (SMAS) are lacking. Thus, the present study aimed to provide new conclusive data about the topography, density, and relationship of the SMAS blood vessels with other components, namely, the fibrous connective tissue and muscles. Methods: The study included a control lot of 42 cases from the archive of the radiology department. In this group, nuclear magnetic resonance angiography (MRA) was performed in order to identify the main sources of vascular supply. In the second group, tissue samples were collected from the midfacial region of 45 patients from the Oro-Maxillo-Facial and Plastic and Reconstructive Surgery clinics of 'St. Spiridon' County Clinical Emergency Hospital, Iasi. These patients received surgery for excision of tumoral formations that did not involve SMAS components. These samples underwent micro-CT analysis, hematoxylin and eosin (HE) staining, as well as immunohistochemical (IHC) staining for collagen type III, muscle tissue, and the vascular endothelium. Results: We discovered the particular way in which the SMAS components interrelate with vascularization and the regional differences between them. We have discovered a new vascular network specific to the SMAS, highlighted by both the micro-CT technique and microscopy on slides with special IHC staining. Significant differences were observed in the topographic arrangement, density, and relationships of the microscopic vasculature across midfacial regions. IHC staining provided morphological and functional information about the structure and vascularization of SMAS. Conclusions: The MRA technique could not detect the structural blood vessels of the SMAS and other methods for their in vivo visualization must be sought. The blood vessels of the SMAS mainly follow the topography of the muscle fibers. From the SMAS layer where they are found, the distribution branches reach the stroma of the region and the hypoderm. Our data can contribute to the development of surgical techniques tailored to each individual patient, as well as the enhancement of methods for stimulating cutaneous angiogenesis, improving scarring in this region, and advancing biotissue engineering techniques.
Collapse
Affiliation(s)
- Delia Hînganu
- Department of Morpho-Functional Sciences I, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.H.); (L.L.)
| | - Marius Valeriu Hînganu
- Department of Morpho-Functional Sciences I, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.H.); (L.L.)
| | - Camelia Tamaș
- Department of Plastic and Reconstructive Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Victor Vlad Costan
- Department of Oral and Maxillo-Facial Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (V.V.C.); (R.P.C.)
| | - Liliana Hristian
- Department of Engineering and Design of Textile Products, “Gheorghe Asachi” Technical University of Iasi, 700050 Iasi, Romania;
| | - Dragoș Negru
- Department of Radiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Anca Elena Calistru
- Department of Pedotechnics, “Ion Ionescu de la Brad” University of Life Sciences, 700490 Iasi, Romania;
| | - Ramona Paula Cucu
- Department of Oral and Maxillo-Facial Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (V.V.C.); (R.P.C.)
| | - Ludmila Lozneanu
- Department of Morpho-Functional Sciences I, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.H.); (L.L.)
| |
Collapse
|
6
|
Imran M, Moyle PM, Kamato D, Mohammed Y. Advances in, and prospects of, 3D preclinical models for skin drug discovery. Drug Discov Today 2024; 29:104208. [PMID: 39396673 DOI: 10.1016/j.drudis.2024.104208] [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: 05/20/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
The skin has an important role in regulating homeostasis and protecting the body from endogenous and exogenous microenvironments. Although 3D models for drug discovery have been extensively studied, there is a growing demand for more advanced 3D skin models to enhance skin research. The use of these advanced skin models holds promise across domains such as cosmetics, skin disease treatments, and toxicity testing of new therapeutics. Recent advances include the development of skin-on-a-chip, spheroids, reconstructed skin, organoids, and computational approaches, including quantitative structure-activity relationship (QSAR) and quantitative structure-property relationship (QSPR) research. These innovations are bridging the gap between traditional 2D and advanced 3D models, moving progress from research to clinical applications. In this review, we highlight in vitro and computational skin models with advanced drug discovery for skin-related applications.
Collapse
Affiliation(s)
- Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Peter Michael Moyle
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia; School of Environment and Science, Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD 4111, Australia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| |
Collapse
|
7
|
Akbarian M, Kianpour M, Tayebi L. Fabricating Multiphasic Angiogenic Scaffolds Using Amyloid/Roxadustat-Assisted High-Temperature Protein Printing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36983-37006. [PMID: 38953207 DOI: 10.1021/acsami.4c06207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Repairing multiphasic defects is cumbersome. This study presents new soft and hard scaffold designs aimed at facilitating the regeneration of multiphasic defects by enhancing angiogenesis and improving cell attachment. Here, the nonimmunogenic, nontoxic, and cost-effective human serum albumin (HSA) fibril (HSA-F) was used to fabricate thermostable (up to 90 °C) and hard printable polymers. Additionally, using a 10.0 mg/mL HSA-F, an innovative hydrogel was synthesized in a mixture with 2.0% chitosan-conjugated arginine, which can gel in a cell-friendly and pH physiological environment (pH 7.4). The presence of HSA-F in both hard and soft scaffolds led to an increase in significant attachment of the scaffolds to the human periodontal ligament fibroblast (PDLF), human umbilical vein endothelial cell (HUVEC), and human osteoblast. Further studies showed that migration (up to 157%), proliferation (up to 400%), and metabolism (up to 210%) of these cells have also improved in the direction of tissue repair. By examining different in vitro and ex ovo experiments, we observed that the final multiphasic scaffold can increase blood vessel density in the process of per-vascularization as well as angiogenesis. By providing a coculture environment including PDLF and HUVEC, important cross-talk between these two cells prevails in the presence of roxadustat drug, a proangiogenic in this study. In vitro and ex ovo results demonstrated significant enhancements in the angiogenic response and cell attachment, indicating the effectiveness of the proposed design. This approach holds promise for the regeneration of complex tissue defects by providing a conducive environment for vascularization and cellular integration, thus promoting tissue healing.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| | - Maryam Kianpour
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| |
Collapse
|
8
|
Rajeev A, Kansara K, Bhatia D. Navigating the challenges and exploring the perspectives associated with emerging novel biomaterials. Biomater Sci 2024; 12:3565-3581. [PMID: 38832912 DOI: 10.1039/d4bm00376d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The field of biomaterials is a continuously evolving interdisciplinary field encompassing biological sciences, materials sciences, chemical sciences, and physical sciences with a multitude of applications realized every year. However, different biomaterials developed for different applications have unique challenges in the form of biological barriers, and addressing these challenges simultaneously is also a challenge. Nevertheless, immense progress has been made through the development of novel materials with minimal adverse effects such as DNA nanostructures, specific synthesis strategies based on supramolecular chemistry, and modulating the shortcomings of existing biomaterials through effective functionalization techniques. This review discusses all these aspects of biomaterials, including the challenges at each level of their development and application, proposed countermeasures for these challenges, and some future directions that may have potential benefits.
Collapse
Affiliation(s)
- Ashwin Rajeev
- Department of Biosciences and Bioengineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382355, India.
| | - Krupa Kansara
- Department of Biosciences and Bioengineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382355, India.
| | - Dhiraj Bhatia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382355, India.
| |
Collapse
|
9
|
Da Silva D, Crous A, Abrahamse H. Enhancing Osteoblast Differentiation from Adipose-Derived Stem Cells Using Hydrogels and Photobiomodulation: Overcoming In Vitro Limitations for Osteoporosis Treatment. Curr Issues Mol Biol 2024; 46:6346-6365. [PMID: 39057021 PMCID: PMC11276038 DOI: 10.3390/cimb46070379] [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: 05/02/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoporosis represents a widespread and debilitating chronic bone condition that is increasingly prevalent globally. Its hallmark features include reduced bone density and heightened fragility, which significantly elevate the risk of fractures due to the decreased presence of mature osteoblasts. The limitations of current pharmaceutical therapies, often accompanied by severe side effects, have spurred researchers to seek alternative strategies. Adipose-derived stem cells (ADSCs) hold considerable promise for tissue repair, albeit they encounter obstacles such as replicative senescence in laboratory conditions. In comparison, employing ADSCs within three-dimensional (3D) environments provides an innovative solution, replicating the natural extracellular matrix environment while offering a controlled and cost-effective in vitro platform. Moreover, the utilization of photobiomodulation (PBM) has emerged as a method to enhance ADSC differentiation and proliferation potential by instigating cellular stimulation and facilitating beneficial performance modifications. This literature review critically examines the shortcomings of current osteoporosis treatments and investigates the potential synergies between 3D cell culture and PBM in augmenting ADSC differentiation towards osteogenic lineages. The primary objective of this study is to assess the efficacy of combined 3D environments and PBM in enhancing ADSC performance for osteoporosis management. This research is notably distinguished by its thorough scrutiny of the existing literature, synthesis of recent advancements, identification of future research trajectories, and utilization of databases such as PubMed, Scopus, Web of Science, and Google Scholar for this literature review. Furthermore, the exploration of biomechanical and biophysical stimuli holds promise for refining treatment strategies. The future outlook suggests that integrating PBM with ADSCs housed within 3D environments holds considerable potential for advancing bone regeneration efforts. Importantly, this review aspires to catalyse further advancements in combined therapeutic strategies for osteoporosis regeneration.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Johannesburg 2028, South Africa; (D.D.S.); (A.C.)
| |
Collapse
|
10
|
Wang Z, Tuerxun P, Ng T, Yan Y, Zhao K, Jian Y, Jia X. Enhancing angiogenesis in peri-implant soft tissue with bioactive silk fibroin microgroove coatings on zirconia surfaces. Regen Biomater 2024; 11:rbae068. [PMID: 39027360 PMCID: PMC11257716 DOI: 10.1093/rb/rbae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/12/2024] [Accepted: 06/02/2024] [Indexed: 07/20/2024] Open
Abstract
Zirconia abutments and restorations have improved the aesthetic appeal of implant restoration, yet peri-implantitis poses a significant threat to long-term success. The soft tissue surrounding implants is a crucial biological barrier against inflammation and subsequent bone loss. Peri-implantitis, akin to periodontitis, progresses rapidly and causes extensive tissue damage. Variations in tissue structure significantly influence disease progression, particularly the lower vascular density in peri-implant connective tissue, compromising its ability to combat infection and provide essential nutrients. Blood vessels within this tissue are vital for healing, with angiogenesis playing a key role in immune defense and tissue repair. Enhancing peri-implant soft tissue angiogenesis holds promise for tissue integration and inflammation control. Microgroove surfaces have shown potential in guiding vessel growth, but using subtractive technologies to carve microgrooves on zirconia surfaces may compromise mechanical integrity. In this study, we utilized inkjet printing to prepare bioactive silk fibroin microgrooves (SFMG) coating with different sizes on zirconia surfaces. SFMG coating, particularly with 90 µm width and 10 µm depth, effectively directed human umbilical vein endothelial cells (HUVECs) along microgrooves, promoting their proliferation, migration, and tube formation. The expression of vascular endothelial growth factor A and fibroblast growth factor in HUVECs growing on SFMG coating was upregulated. Additionally, the SFMG coating activated the PI3K-AKT pathway and increased glycolytic enzyme gene expression in HUVECs. In conclusion, SFMG coating enhances HUVEC growth and angiogenesis potential by activating the PI3K-AKT pathway and glycolysis, showing promise for improving tissue integration and mitigating inflammation in zirconia abutments and restorations.
Collapse
Affiliation(s)
- Zhihan Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Palati Tuerxun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Takkun Ng
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yinuo Yan
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Ke Zhao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yutao Jian
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xiaoshi Jia
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
11
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024:10.1007/s10456-024-09928-6. [PMID: 38842751 DOI: 10.1007/s10456-024-09928-6] [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: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Wei SY, Chen PY, Tsai MC, Hsu TL, Hsieh CC, Fan HW, Chen TH, Xie RH, Chen GY, Chen YC. Enhancing the Repair of Substantial Volumetric Muscle Loss by Creating Different Levels of Blood Vessel Networks Using Pre-Vascularized Nerve Hydrogel Implants. Adv Healthc Mater 2024; 13:e2303320. [PMID: 38354361 DOI: 10.1002/adhm.202303320] [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: 09/29/2023] [Revised: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Volumetric muscle loss (VML), a severe muscle tissue loss from trauma or surgery, results in scarring, limited regeneration, and significant fibrosis, leading to lasting reductions in muscle mass and function. A promising approach for VML recovery involves restoring vascular and neural networks at the injury site, a process not extensively studied yet. Collagen hydrogels have been investigated as scaffolds for blood vessel formation due to their biocompatibility, but reconstructing blood vessels and guiding innervation at the injury site is still difficult. In this study, collagen hydrogels with varied densities of vessel-forming cells are implanted subcutaneously in mice, generating pre-vascularized hydrogels with diverse vessel densities (0-145 numbers/mm2) within a week. These hydrogels, after being transplanted into muscle injury sites, are assessed for muscle repair capabilities. Results showed that hydrogels with high microvessel densities, filling the wound area, effectively reconnected with host vasculature and neural networks, promoting neovascularization and muscle integration, and addressing about 63% of the VML.
Collapse
Affiliation(s)
- Shih-Yen Wei
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| | - Po-Yu Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| | - Min-Chun Tsai
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| | - Ting-Lun Hsu
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| | - Chia-Chang Hsieh
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| | - Hsiu-Wei Fan
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| | - Tzu-Hsuan Chen
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, 15289, USA
| | - Ren-Hao Xie
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300193, Taiwan
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300193, Taiwan
| | - Guan-Yu Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300193, Taiwan
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300193, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300193, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, 300193, Taiwan
| | - Ying-Chieh Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, 300044, Taiwan
| |
Collapse
|
13
|
Anitua E, Troya M, Zalduendo M, Tierno R, Alkhraisat MH, Osinalde N, Fullaondo A, Zubiaga AM. Improving the mechanical and biological functions of cell sheet constructs: The interplay of human-derived periodontal ligament stem cells, endothelial cells and plasma rich in growth factors. Biomed Pharmacother 2024; 174:116599. [PMID: 38640711 DOI: 10.1016/j.biopha.2024.116599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE The aim of this study was to produce and characterize triple-layered cell sheet constructs with varying cell compositions combined or not with the fibrin membrane scaffold obtained by the technology of Plasma Rich in Growth Factors (mPRGF). MATERIALS AND METHODS Human primary cultures of periodontal ligament stem cells (hPDLSCs) were isolated, and their stemness nature was evaluated. Three types of triple-layered composite constructs were generated, composed solely of hPDLSCs or combined with human umbilical vein endothelial cells (HUVECs), either as a sandwiched endothelial layer or as coculture sheets of both cell phenotypes. These three triple-layered constructs were also manufactured using mPRGF as cell sheets' support. Necrosis, glucose consumption, secretion of extracellular matrix proteins and synthesis of proangiogenic factors were determined. Histological evaluations and proteomic analyses were also performed. RESULTS The inclusion of HUVECs did not clearly improve the properties of the multilayered constructs and yet hindered their optimal conformation. The presence of mPRGF prevented the shrinkage of cell sheets, stimulated the metabolic activity and increased the matrix synthesis. At the proteome level, mPRGF conferred a dramatic advantage to the hPDLSC constructs in their ability to provide a suitable environment for tissue regeneration by inducing the expression of proteins necessary for bone morphogenesis and cellular proliferation. CONCLUSIONS hPDLSCs' triple-layer construct onto mPRGF emerges as the optimal structure for its use in regenerative therapeutics. CLINICAL RELEVANCE These results suggest the suitability of mPRGF as a promising tool to support cell sheet formation by improving their handling and biological functions.
Collapse
Affiliation(s)
- Eduardo Anitua
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| | - María Troya
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Mar Zalduendo
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Roberto Tierno
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Mohammad H Alkhraisat
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Asier Fullaondo
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Ana M Zubiaga
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
14
|
Iravani S, Nazarzadeh Zare E, Makvandi P. Multifunctional MXene-Based Platforms for Soft and Bone Tissue Regeneration and Engineering. ACS Biomater Sci Eng 2024; 10:1892-1909. [PMID: 38466909 DOI: 10.1021/acsbiomaterials.3c01770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
MXenes and their composites hold great promise in the field of soft and bone tissue regeneration and engineering (TRE). However, there are challenges that need to be overcome, such as ensuring biocompatibility and controlling the morphologies of MXene-based scaffolds. The future prospects of MXenes in TRE include enhancing biocompatibility through surface modifications, developing multifunctional constructs, and conducting in vivo studies for clinical translation. The purpose of this perspective about MXenes and their composites in soft and bone TRE is to critically evaluate their potential applications and contributions in this field. This perspective aims to provide a comprehensive analysis of the challenges, advantages, limitations, and future prospects associated with the use of MXenes and their composites for soft and bone TRE. By examining the existing literature and research, the review seeks to consolidate the current knowledge and highlight the key findings and advancements in MXene-based TRE. It aims to contribute to the understanding of MXenes' role in promoting soft and bone TRE, addressing the challenges faced in terms of biocompatibility, morphology control, and tissue interactions.
Collapse
Affiliation(s)
- Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Avenue, Isfahan 81756-33551, Iran
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan 36716-45667, Iran
- Centre of Research Impact and Outreach, Chitkara University, Rajpura 140417, Punjab, India
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, Zhejiang, China
- Chitkara Centre for Research and Development, Chitkara University, Kalujhanda 174103, Himachal Pradesh, India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| |
Collapse
|
15
|
Lallo V, Bracaglia LG. Influencing Endothelial Cells' Roles in Inflammation and Wound Healing Through Nucleic Acid Delivery. Tissue Eng Part A 2024; 30:272-286. [PMID: 38149606 DOI: 10.1089/ten.tea.2023.0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
Tissue engineering and wound-healing interventions are often designed for use in diseased and inflamed environments. In this space, endothelial cells (ECs) are crucial regulators of inflammation and healing, as they are the primary contact for recruitment of immune cells, as well as production of proinflammatory cytokines, which can stimulate or reduce inflammation. Alternatively, proliferation and spreading of ECs result in the formation of new vascular tissue or repair of damaged tissue, both critical for wound healing. Targeting ECs with specific nucleic acids could reduce unwanted inflammation or promote tissue regeneration as needed, which are two large issues involved in many regenerative medicine goals. Polymeric delivery systems are tools that can control the delivery of nucleic acids and prolong their effects. This review describes the use of polymeric vehicles for the delivery of nucleic acids to ECs for tissue engineering. Impact statement Tissue engineering is a rapidly growing field that has the potential to resolve many disease states and improve the quality of life of patients. In some applications, tissue-engineered strategies or constructs are developed to rebuild spaces damaged by disease or degeneration. To rebuild the native tissue, these constructs may need to interact with unwanted immune activity and cells. Various immune cells are often the focus of therapies as they are critical players in the inflammatory response; however, endothelial cells are also an extremely important and promising target in these cases. In addition, controlled delivery of specific-acting molecules, such as nucleic acids, is of growing interest for the regeneration and health of a variety of different tissues. It is important to understand what has been done and the potential of these targets and therapeutics for future investigation and advancements in tissue engineering.
Collapse
Affiliation(s)
- Valerie Lallo
- Department of Chemical and Biological Engineering, Villanova University, Villanova, Pennsylvania, USA
| | - Laura G Bracaglia
- Department of Chemical and Biological Engineering, Villanova University, Villanova, Pennsylvania, USA
| |
Collapse
|
16
|
Minne M, Terrie L, Wüst R, Hasevoets S, Vanden Kerchove K, Nimako K, Lambrichts I, Thorrez L, Declercq H. Generating human skeletal myoblast spheroids for vascular myogenic tissue engineering. Biofabrication 2024; 16:025035. [PMID: 38437715 DOI: 10.1088/1758-5090/ad2fd5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/04/2024] [Indexed: 03/06/2024]
Abstract
Engineered myogenic microtissues derived from human skeletal myoblasts offer unique opportunities for varying skeletal muscle tissue engineering applications, such asin vitrodrug-testing and disease modelling. However, more complex models require the incorporation of vascular structures, which remains to be challenging. In this study, myogenic spheroids were generated using a high-throughput, non-adhesive micropatterned surface. Since monoculture spheroids containing human skeletal myoblasts were unable to remain their integrity, co-culture spheroids combining human skeletal myoblasts and human adipose-derived stem cells were created. When using the optimal ratio, uniform and viable spheroids with enhanced myogenic properties were achieved. Applying a pre-vascularization strategy, through addition of endothelial cells, resulted in the formation of spheroids containing capillary-like networks, lumina and collagen in the extracellular matrix, whilst retaining myogenicity. Moreover, sprouting of endothelial cells from the spheroids when encapsulated in fibrin was allowed. The possibility of spheroids, from different maturation stages, to assemble into a more large construct was proven by doublet fusion experiments. The relevance of using three-dimensional microtissues with tissue-specific microarchitecture and increased complexity, together with the high-throughput generation approach, makes the generated spheroids a suitable tool forin vitrodrug-testing and human disease modeling.
Collapse
Affiliation(s)
- Mendy Minne
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Lisanne Terrie
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Rebecca Wüst
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Steffie Hasevoets
- Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, UHasselt, Diepenbeek, Belgium
| | - Kato Vanden Kerchove
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Kakra Nimako
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Ivo Lambrichts
- Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, UHasselt, Diepenbeek, Belgium
| | - Lieven Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| | - Heidi Declercq
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven campus KULAK, Kortrijk, Belgium
| |
Collapse
|
17
|
Zhu J, Zhao F, Chai Y, Jia X, Li F. Evaluating the Efficacy of Dedifferentiated Fat Cells (DFATs) vs Adipose-Derived Stem Cells (ASCs) in Enhancing the Viability of Fat Grafts. Aesthet Surg J 2024; 44:NP307-NP318. [PMID: 37943807 DOI: 10.1093/asj/sjad342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Enhancing graft fat survival remains a paramount challenge in autologous fat transplantation surgeries. Dedifferentiated fat cells (DFATs) and adipose-derived stem cells (ASCs) represent 2 pivotal cells with potential to improve fat graft survival rates. OBJECTIVES In this study we aimed to compare the effectiveness of DFATs and ASCs in promoting fat graft survival, emphasizing their adipogenic and angiogenic capabilities. METHODS Both in vitro and in vivo experiments were conducted. In vitro assessments compared adipogenesis, angiogenesis, osteogenesis, chondrogenesis, cell migration abilities, and surface markers. For in vivo evaluation, a cell-assisted lipotransfer animal model was employed to gauge graft volume retention and histological morphology. Analysis techniques included hematoxylin and eosin staining, Western blotting, and real-time polymerase chain reaction. RESULTS In vitro findings suggested a slight superiority of DFATs in adipogenesis and angiogenesis compared to ASCs. In vivo tests demonstrated both cell types surpassed the control in terms of graft volume retention, with the DFATs group marginally outperforming in retention rates and the ASC group presenting a slightly enhanced graft tissue structure. CONCLUSIONS Our study underscores the distinct advantages of DFATs and ASCs in bolstering fat graft survival, offering potentially novel insights for plastic surgeons aiming to elevate fat graft survival rates.
Collapse
|
18
|
Norberg AE, Bakirci E, Lim KS, Dalton PD, Woodfield TBF, Lindberg GCJ. Bioassembly of hemoglobin-loaded photopolymerizable spheroids alleviates hypoxia-induced cell death. Biofabrication 2024; 16:025026. [PMID: 38373325 DOI: 10.1088/1758-5090/ad2a7d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 02/21/2024]
Abstract
The delivery of oxygen within tissue engineered constructs is essential for cell survivability; however, achieving this within larger biofabricated constructs poses a significant challenge. Efforts to overcome this limitation often involve the delivery of synthetic oxygen generating compounds. The application of some of these compounds is problematic for the biofabrication of living tissues due to inherent issues such as cytotoxicity, hyperoxia and limited structural stability due to oxygen inhibition of radical-based crosslinking processes. This study aims to develop an oxygen delivering system relying on natural-derived components which are cytocompatible, allow for photopolymerization and advanced biofabrication processes, and improve cell survivability under hypoxia (1% O2). We explore the binding of human hemoglobin (Hb) as a natural oxygen deposit within photopolymerizable allylated gelatin (GelAGE) hydrogels through the spontaneous complex formation of Hb with negatively charged biomolecules (heparin, hyaluronic acid, and bovine serum albumin). We systematically study the effect of biomolecule inclusion on cytotoxicity, hydrogel network properties, Hb incorporation efficiency, oxygen carrying capacity, cell viability, and compatibility with 3D-bioassembly processes within melt electrowritten (MEW) scaffolds. All biomolecules were successfully incorporated within GelAGE hydrogels, displaying controllable mechanical properties and cytocompatibility. Results demonstrated efficient and tailorable Hb incorporation within GelAGE-Heparin hydrogels. The developed system was compatible with microfluidics and photopolymerization processes, allowing for the production of GelAGE-Heparin-Hb spheres. Hb-loaded spheres were assembled into MEW polycaprolactone scaffolds, significantly increasing the local oxygen levels. Ultimately, cells within Hb-loaded constructs demonstrated good cell survivability under hypoxia. Taken together, we successfully developed a hydrogel system that retains Hb as a natural oxygen deposit post-photopolymerization, protecting Hb from free-radical oxidation while remaining compatible with biofabrication of large constructs. The developed GelAGE-Heparin-Hb system allows for physoxic oxygen delivery and thus possesses a vast potential for use across broad tissue engineering and biofabrication strategies to help eliminate cell death due to hypoxia.
Collapse
Affiliation(s)
- Axel E Norberg
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Khoon S Lim
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Paul D Dalton
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Tim B F Woodfield
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Gabriella C J Lindberg
- Dept of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| |
Collapse
|
19
|
Biniazan F, Stoian A, Haykal S. Adipose-Derived Stem Cells: Angiogenetic Potential and Utility in Tissue Engineering. Int J Mol Sci 2024; 25:2356. [PMID: 38397032 PMCID: PMC10889096 DOI: 10.3390/ijms25042356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Adipose tissue (AT) is a large and important energy storage organ as well as an endocrine organ with a critical role in many processes. Additionally, AT is an enormous and easily accessible source of multipotent cell types used in our day for all types of tissue regeneration. The ability of adipose-derived stem cells (ADSCs) to differentiate into other types of cells, such as endothelial cells (ECs), vascular smooth muscle cells, or cardiomyocytes, is used in tissue engineering in order to promote/stimulate the process of angiogenesis. Being a key for future successful clinical applications, functional vascular networks in engineered tissue are targeted by numerous in vivo and ex vivo studies. The article reviews the angiogenic potential of ADSCs and explores their capacity in the field of tissue engineering (TE).
Collapse
Affiliation(s)
- Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Toronto, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada
| |
Collapse
|
20
|
Lackner F, Šurina P, Fink J, Kotzbeck P, Kolb D, Stana J, Grab M, Hagl C, Tsilimparis N, Mohan T, Stana Kleinschek K, Kargl R. 4-Axis 3D-Printed Tubular Biomaterials Imitating the Anisotropic Nanofiber Orientation of Porcine Aortae. Adv Healthc Mater 2024; 13:e2302348. [PMID: 37807640 PMCID: PMC11469240 DOI: 10.1002/adhm.202302348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/16/2023] [Indexed: 10/10/2023]
Abstract
Many of the peculiar properties of the vasculature are related to the arrangement of anisotropic proteinaceous fibers in vessel walls. Understanding and imitating these arrangements can potentially lead to new therapies for cardiovascular diseases. These can be pre-surgical planning, for which patient-specific ex vivo anatomical models for endograft testing are of interest. Alternatively, therapies can be based on tissue engineering, for which degradable in vitro cell growth substrates are used to culture replacement parts. In both cases, materials are desirable that imitate the biophysical properties of vessels, including their tubular shapes and compliance. This work contributes to these demands by offering methods for the manufacturing of anisotropic 3D-printed nanofibrous tubular structures that have similar biophysical properties as porcine aortae, that are biocompatible, and that allow for controlled nutrient diffusion. Tubes of various sizes with axial, radial, or alternating nanofiber orientation along the blood flow direction are manufactured by a customized method. Blood pressure-resistant, compliant, stable, and cell culture-compatible structures are obtained, that can be degraded in vitro on demand. It is suggested that these healthcare materials can contribute to the next generation of cardiovascular therapies of ex vivo pre-surgical planning or in vitro cell culture.
Collapse
Affiliation(s)
- Florian Lackner
- Institute for Chemistry and Technology of Biobased System (IBioSys)Graz University of TechnologyStremayrgasse 98010GrazAustria
| | - Paola Šurina
- Institute for Chemistry and Technology of Biobased System (IBioSys)Graz University of TechnologyStremayrgasse 98010GrazAustria
| | - Julia Fink
- COREMED ‐ Centre of Regenerative and Precision MedicineJOANNEUM RESEARCH Forschungsgesellschaft mbHNeue Stiftingtalstraße 28010GrazAustria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of SurgeryMedical University of GrazAuenbruggerplatz 29/48036GrazAustria
| | - Petra Kotzbeck
- COREMED ‐ Centre of Regenerative and Precision MedicineJOANNEUM RESEARCH Forschungsgesellschaft mbHNeue Stiftingtalstraße 28010GrazAustria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of SurgeryMedical University of GrazAuenbruggerplatz 29/48036GrazAustria
| | - Dagmar Kolb
- Core Unit Ultrastructure AnalysisMedical University GrazStiftingtalstraße 6/II8010GrazAustria
- Gottfried Schatz Research Center for Cell Signaling Metabolism and AgingMedical University GrazStiftingtalstraße 68010GrazAustria
| | - Jan Stana
- Department of Vascular SurgeryLudwig Maximilian University MunichMarchioninistraße 1581377MunichGermany
| | - Maximilian Grab
- Department of Cardiac SurgeryLudwig Maximilian University MunichMarchioninistraße 1581377MunichGermany
| | - Christian Hagl
- Department of Cardiac SurgeryLudwig Maximilian University MunichMarchioninistraße 1581377MunichGermany
| | - Nikolaos Tsilimparis
- Department of Vascular SurgeryLudwig Maximilian University MunichMarchioninistraße 1581377MunichGermany
| | - Tamilselvan Mohan
- Institute for Chemistry and Technology of Biobased System (IBioSys)Graz University of TechnologyStremayrgasse 98010GrazAustria
- Laboratory for Characterization and Processing of PolymersUniversity of MariborSmetanova ulica 16Maribor2000Slovenia
| | - Karin Stana Kleinschek
- Institute for Chemistry and Technology of Biobased System (IBioSys)Graz University of TechnologyStremayrgasse 98010GrazAustria
| | - Rupert Kargl
- Institute for Chemistry and Technology of Biobased System (IBioSys)Graz University of TechnologyStremayrgasse 98010GrazAustria
- Laboratory for Characterization and Processing of PolymersUniversity of MariborSmetanova ulica 16Maribor2000Slovenia
| |
Collapse
|
21
|
Jia S, Wang X, Wang G, Wang X. Mechanism and application of β-adrenoceptor blockers in soft tissue wound healing. Med Res Rev 2024; 44:422-452. [PMID: 37470332 DOI: 10.1002/med.21984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 06/01/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
Soft tissue damage stimulates sympathetic nerves to release large amounts of catecholamine hormones which bind to β-adrenergic receptors (β-ARs) on the cell membrane surface. It activates the downstream effector molecules and impairs soft tissue wound healing. β-blockers specifically inhibit β-ARs activation in acute/chronic skin lesions and ulcerative hemangiomas. They also accelerate soft tissue wound healing by shortening the duration of inflammation, speeding keratinocyte migration and reepithelialization, promoting wound contraction and angiogenesis, and inhibiting bacterial virulence effects. In addition, β-blockers shorten wound healing periods in patients with severe thermal damage by reducing the hypermetabolic response. While β-blockers promote/inhibit corneal epithelial cell regeneration and restores limbal stem/progenitor cells function, it could well accelerate/delay corneal wound healing. Given these meaningful effects, a growing number of studies are focused on examining the efficacy and safety of β-blockers in soft tissue wound repair, including acute and chronic wounds, severe thermal damage, ulcerated infantile hemangioma, corneal wounds, and other soft tissue disorders. However, an intensive investigation on their acting mechanisms is imperatively needed. The purpose of this article is to summerize the roles of β-blockers in soft tissue wound healing and explore their clinical applications.
Collapse
Affiliation(s)
- Shasha Jia
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
- School of Stomatology, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Xueya Wang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
- School of Stomatology, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Guowei Wang
- Department of Stomatology, No. 971 Hospital of the Chinese Navy, Qingdao, Shandong, People's Republic of China
| | - Xiaojing Wang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
- School of Stomatology, Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
22
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
23
|
Dasgupta S, Reddy KP, Datta P, Barui A. Vitamin D3-incorporated chitosan/collagen/fibrinogen scaffolds promote angiogenesis and endothelial transition via HIF-1/IGF-1/VEGF pathways in dental pulp stem cells. Int J Biol Macromol 2023; 253:127325. [PMID: 37820916 DOI: 10.1016/j.ijbiomac.2023.127325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Effective vascularization during wound healing remains a critical challenge in the regeneration of skin tissue. On the other hand, mesenchymal stem cell (MSC) to endothelial phenotype transition (MEnDoT) is a potential phenomenon grossly underexplored in vascularized skin tissue engineering. Vitamin D3 has a proven role in promoting MEnDoT. Hence, a D3-incorporated scaffold made with biocompatible materials such as chitosan, collagen and fibrinogen should be able to promote endothelial lineage transition in vitro for tissue engineering purposes. In this study, we developed vitamin D3 incorporated chitosan-collagen-fibrinogen (CCF-D3) scaffolds physically crosslinked under UV and conducted thorough physicochemical and biological assays on it compared to a control scaffold without vitamin D3. Our study for the first time reports the potential vascularization property of the CCF-D3 scaffold by inducing the transitions of dental pulp MSC to endothelial lineage via the HIF-1/IGF-1/VEGF pathways. MSC seeded on UV-exposed CCF-D3 scaffolds had higher cell viability and transitioned towards endothelial lineage was observed by elevated proliferative and endothelial-specific gene expressions and flow cytometric analysis of SCA-1+ antibody. The difference in VEGF-A and α-SMA expressions was also observed in the D3-CCF scaffold compared to the scaffolds without D3.
Collapse
Affiliation(s)
- Shalini Dasgupta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, India
| | | | - Pallab Datta
- National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, India.
| |
Collapse
|
24
|
Wei SY, Chen PY, Hsieh CC, Chen YS, Chen TH, Yu YS, Tsai MC, Xie RH, Chen GY, Yin GC, Melero-Martin JM, Chen YC. Engineering large and geometrically controlled vascularized nerve tissue in collagen hydrogels to restore large-sized volumetric muscle loss. Biomaterials 2023; 303:122402. [PMID: 37988898 DOI: 10.1016/j.biomaterials.2023.122402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Developing scalable vascularized and innervated tissue is a critical challenge for the successful clinical application of tissue-engineered constructs. Collagen hydrogels are extensively utilized in cell-mediated vascular network formation because of their naturally excellent biological properties. However, the substantial increase in hydrogel contraction induced by populated cells limits their long-term use. Previous studies attempted to mitigate this issue by concentrating collagen pre-polymer solutions or synthesizing covalently crosslinked collagen hydrogels. However, these methods only partially reduce hydrogel contraction while hindering blood vessel formation within the hydrogels. To address this challenge, we introduced additional support in the form of a supportive spacer to counteract the contraction forces of populated cells and prevent hydrogel contraction. This approach was found to promote cell spreading, resist hydrogel contraction, control hydrogel/tissue geometry, and even facilitate the engineering of functional blood vessels and host nerve growth in just one week. Subsequently, implanting these engineered tissues into muscle defect sites resulted in timely anastomosis with the host vasculature, leading to enhanced myogenesis, increased muscle innervation, and the restoration of injured muscle functionality. Overall, this innovative strategy expands the applicability of collagen hydrogels in fabricating large vascularized nerve tissue constructs for repairing volumetric muscle loss (∼63 %) and restoring muscle function.
Collapse
Affiliation(s)
- Shih-Yen Wei
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Po-Yu Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Chia-Chang Hsieh
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Yu-Shan Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Tzu-Hsuan Chen
- Department of Materials Science and Engineering, Carnegie Mellon University, PA, USA
| | - Yu-Shan Yu
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Chun Tsai
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Ren-Hao Xie
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Guan-Yu Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Gung-Chian Yin
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ying-Chieh Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan.
| |
Collapse
|
25
|
Kakkad J, Deshmukh P, Gaurkar S. Cartilage's Contribution in Otology: A Comprehensive Review of Its Role in Ear Surgery. Cureus 2023; 15:e49800. [PMID: 38161551 PMCID: PMC10757830 DOI: 10.7759/cureus.49800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
This comprehensive review thoroughly examines the pivotal role of cartilage in otologic surgery, elucidating its multifaceted contributions to both cosmetic and functional outcomes. From reconstructing the external ear to reinforcing the tympanic membrane and restoring the ossicular chain, cartilage emerges as a versatile and resilient biological material with unique properties that make it an invaluable resource for otologic surgeons. The review explores the nuances of cartilage's applications in various surgical contexts, emphasizing its significance in promoting tissue regeneration and healing. The text delves into advancements in tissue engineering, biodegradable scaffolds, and 3D printing technology, pointing toward a future where more precise and personalized interventions may redefine the landscape of otologic surgery. The convergence of these innovations holds the promise of elevating the standard of care, minimizing complications, and improving the quality of life for patients undergoing cartilage-based otologic procedures. This synthesis of current knowledge and future possibilities provides a valuable resource for otologists, surgeons, and researchers in the dynamic field of otology.
Collapse
Affiliation(s)
- Jasleen Kakkad
- Otolaryngology, Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prasad Deshmukh
- Otolaryngology, Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sagar Gaurkar
- Otolaryngology, Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
26
|
Liang J, Zhao J, Chen Y, Li B, Li Y, Lu F, Dong Z. New Insights and Advanced Strategies for In Vitro Construction of Vascularized Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:692-709. [PMID: 37409413 DOI: 10.1089/ten.teb.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Inadequate vascularization is a significant barrier to clinical application of large-volume tissue engineered grafts. In contrast to in vivo vascularization, in vitro prevascularization shortens the time required for host vessels to grow into the graft core and minimizes necrosis in the core region of the graft. However, the challenge of prevascularization is to construct hierarchical perfusable vascular networks, increase graft volume, and form a vascular tip that can anastomose with host vessels. Understanding advances in in vitro prevascularization techniques and new insights into angiogenesis could overcome these obstacles. In the present review, we discuss new perspectives on angiogenesis, the differences between in vivo and in vitro tissue vascularization, the four elements of prevascularized constructs, recent advances in perfusion-based in vitro prevascularized tissue fabrication, and prospects for large-volume prevascularized tissue engineering.
Collapse
Affiliation(s)
- Jiancong Liang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jing Zhao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yunzi Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Bin Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ziqing Dong
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
27
|
Wang YT, Meng XT. A review of the evidence to support electrical stimulation -induced vascularization in engineered tissue. Regen Ther 2023; 24:237-244. [PMID: 37534238 PMCID: PMC10393514 DOI: 10.1016/j.reth.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/25/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023] Open
Abstract
Tissue engineering presents a promising solution for regenerative medicine and the success depends on the supply of oxygen/nutrients to the cells by rapid vascularization. More and more technologies are being developed to facilitate vascularization of engineered tissues. In this review, we indicated that a regulatory system which influences all angiogenesis associated cells to achieve their desired functional state is ideal for the construction of vascularized engineered tissues in vitro. We presented the evidence that electrical stimulation (ES) enhances the synergistic promotion of co-cultured angiogenesis associated cells and its potential regulatory mechanisms, highlighted the potential advantages of a combination of mesenchymal stem cells (MSCs), endothelial cells (ECs) and ES to achieve tissue vascularization, with particular emphasis on the different biological pathways of ES-regulated ECs. Finally, we proposed the future direction of using ES to reconstruct engineered tissue blood vessels, pointed out the potential advantages and disadvantages of ES application on tissue vascularization.
Collapse
Affiliation(s)
- Ying-tong Wang
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiao-ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
28
|
Martin‐Piedra MA, Carmona G, Campos F, Carriel V, Fernández‐González A, Campos A, Cuende N, Garzón I, Gacto P, Alaminos M. Histological assessment of nanostructured fibrin-agarose skin substitutes grafted in burnt patients. A time-course study. Bioeng Transl Med 2023; 8:e10572. [PMID: 38023713 PMCID: PMC10658487 DOI: 10.1002/btm2.10572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 12/01/2023] Open
Abstract
A previously developed fibrin-agarose skin model-UGRSKIN-showed promising clinical results in severely burnt patients. To determine the histological parameters associated to the biocompatibility and therapeutic effects of this model, we carried out a comprehensive structural and ultrastructural study of UGRSKIN grafted in severely burnt patients after 3 months of follow-up. The grafted epidermis was analogue to native human skin from day 30th onward, revealing well-structured strata with well-differentiated keratinocytes expressing CK5, CK8, CK10, claudin, plakoglobin, filaggrin, and involucrin in a similar way to controls, suggesting that the epidermis was able to mature and differentiate very early. Melanocytes and Langerhans cells were found from day 30th onward, together with a basement membrane, abundant hemidesmosomes and lack of rete ridges. At the dermal layer, we found an interface between the grafted skin and the host tissue at day 30th, which tended to disappear with time. The grafted superficial dermis showed a progressive increase in properly-oriented collagen fibers, elastic fibers and proteoglycans, including decorin, similarly to control dermis at day 60-90th of in vivo follow-up. Blood vessels determined by CD31 and SMA expression were more abundant in grafted skin than controls, whereas lymphatic vessels were more abundant at day 90th. These results contribute to shed light on the histological parameters associated to biocompatibility and therapeutic effect of the UGRSKIN model grafted in patients and demonstrate that the bioengineered skin grafted in patients is able to mature and differentiate very early at the epithelial level and after 60-90 days at the dermal level.
Collapse
Affiliation(s)
- Miguel Angel Martin‐Piedra
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Gloria Carmona
- Andalusian Network for the Design and Translation of Advanced Therapies (former Andalusian Initiative for Advanced Therapies) ‐ Fundación Andaluza Progreso y Salud, Junta de Andalucía, Seville, Spain; Andalusian Transplant Coordination, Servicio Andaluz de SaludSevilleSpain
- Doctoral program in BiomedicineUniversity of GranadaGranadaSpain
| | - Fernando Campos
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Víctor Carriel
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Ana Fernández‐González
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
- Andalusian Network for the Design and Translation of Advanced Therapies (former Andalusian Initiative for Advanced Therapies) ‐ Fundación Andaluza Progreso y Salud, Junta de Andalucía, Seville, Spain; Andalusian Transplant Coordination, Servicio Andaluz de SaludSevilleSpain
- Unidad de Producción Celular e Ingeniería TisularHospital Universitario Virgen de las NievesGranadaSpain
| | - Antonio Campos
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Natividad Cuende
- Andalusian Network for the Design and Translation of Advanced Therapies (former Andalusian Initiative for Advanced Therapies) ‐ Fundación Andaluza Progreso y Salud, Junta de Andalucía, Seville, Spain; Andalusian Transplant Coordination, Servicio Andaluz de SaludSevilleSpain
| | - Ingrid Garzón
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | | | - Miguel Alaminos
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| |
Collapse
|
29
|
Kim HN, Elgundi Z, Lin X, Fu L, Tang F, Moh ESX, Jung M, Chandrasekar K, Bartlett-Tomasetig F, Foster C, Packer NH, Whitelock JM, Rnjak-Kovacina J, Lord MS. Engineered short forms of perlecan enhance angiogenesis by potentiating growth factor signalling. J Control Release 2023; 362:184-196. [PMID: 37648081 DOI: 10.1016/j.jconrel.2023.08.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
Growth factors are key molecules involved in angiogenesis, a process critical for tissue repair and regeneration. Despite the potential of growth factor delivery to stimulate angiogenesis, limited clinical success has been achieved with this approach. Growth factors interact with the extracellular matrix (ECM), and particularly heparan sulphate (HS), to bind and potentiate their signalling. Here we show that engineered short forms of perlecan, the major HS proteoglycan of the vascular ECM, bind and signal angiogenic growth factors, including fibroblast growth factor 2 and vascular endothelial growth factor-A. We also show that engineered short forms of perlecan delivered in porous chitosan biomaterial scaffolds promote angiogenesis in a rat full thickness dermal wound model, with the fusion of perlecan domains I and V leading to superior vascularisation compared to native endothelial perlecan or chitosan scaffolds alone. Together, this study demonstrates the potential of engineered short forms of perlecan delivered in chitosan scaffolds as next generation angiogenic therapies which exert biological activity via the potentiation of growth factors.
Collapse
Affiliation(s)
- Ha Na Kim
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Molecular Surface Interaction Laboratory, Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Zehra Elgundi
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiaoting Lin
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lu Fu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Fengying Tang
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Edward S X Moh
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, New South Wales 2109, Australia; School of Natural Science, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - MoonSun Jung
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Keerthana Chandrasekar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Florence Bartlett-Tomasetig
- Katherina Gaus Light Microscopy Facility, Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Candice Foster
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nicolle H Packer
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, New South Wales 2109, Australia; School of Natural Science, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Tyree Institute of Health Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
30
|
Xiao Y, Tian Y, Zhang J, Li Q, Shi W, Huang X. Small intestinal submucosa promotes angiogenesis via the Hippo pathway to improve vaginal repair. BIOMOLECULES & BIOMEDICINE 2023; 23:838-847. [PMID: 37183705 PMCID: PMC10494851 DOI: 10.17305/bb.2023.9052] [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: 03/22/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 05/16/2023]
Abstract
Vaginal reconstruction has incorporated the use of gastrointestinal segments for decades, but the technique is inevitably associated with complications. Tissue-engineering techniques, however, have brought great hope for vaginal reconstruction. This study aimed to evaluate the utility of small intestinal submucosa (SIS) in reconstructing clinically significant large vaginal defects in a porcine model and to investigate the role of the Hippo pathway in the vascular remodeling process. The composition and mechanical properties of SIS were characterized. Full-thickness vaginal defects were established in 10 minipig donors, with 4 cm lengths removed and replaced by an equal sized SIS scaffolds. The neovaginas were subjected to macroscopic, histological, immunohistochemical and molecular evaluations at 4 and 12 weeks after the surgery. Four weeks after the operation, extracellular matrix reorganization and complete coverage of the surface of the luminal matrix by vaginal epithelium were observed, accompanied by the formation of a microvascular network and the regeneration of smooth muscles, albeit disorderly arranged. Twelve weeks after implantation, enhancements were seen in the formation of the multi-layered squamous epithelium, angiogenesis, and large muscle bundle formation in the vagina. Additionally, the expression levels of angiogenesis-related proteins, proliferation-related proteins and Hippo pathway-related proteins in the neovagina were significantly increased. These results indicate that SIS could be used to reconstruct large vaginal defects and that the vascular remodeling process is potentially regulated by the Hippo pathway.
Collapse
Affiliation(s)
- Yanlai Xiao
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Obstetrics and Gynecology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanpeng Tian
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jingkun Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qian Li
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenxin Shi
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xianghua Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
31
|
Barone L, Gallazzi M, Rossi F, Papait R, Raspanti M, Zecca PA, Buonarrivo L, Bassani B, Bernardini G, Bruno A, Gornati R. Human Dental Pulp Mesenchymal Stem Cell-Derived Soluble Factors Combined with a Nanostructured Scaffold Support the Generation of a Vascular Network In Vivo. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2479. [PMID: 37686988 PMCID: PMC10489993 DOI: 10.3390/nano13172479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023]
Abstract
Among all strategies directed at developing new tools to support re-vascularization of damaged tissues, the use of pro-angiogenic soluble factors, derived from mesenchymal stem cells (MSCs), appears a promising approach for regenerative medicine. Here, we compared the feasibility of two devices, generated by coupling soluble factors of human dental pulp mesenchymal stem cells (DPSCs), with a nanostructured scaffold, to support angiogenesis once transplanted in mice. DPSCs were obtained from impacted wisdom tooth removal, usually considered surgical waste material. After 28 days, we verified the presence of active blood vessels inside the scaffold through optical and scansion electron microscopy. The mRNA expression of surface antigens related to macrophage polarization (CD68, CD80, CD86, CD163, CD206), as well as pro-angiogenic markers (CD31, CD34, CD105, Angpt1, Angpt2, CDH5) was evaluated by real-time PCR. Our results demonstrate the capability of DPSC-scaffold and DPSC soluble factors-scaffold to support angiogenesis, similarly to adipose stem cells, whereas the absence of blood vessels was found in the scaffold grafted alone. Our results provide evidence that DPSC-conditioned medium can be proposed as a cell-free preparation able to support angiogenesis, thus, providing a relevant tool to overcome the issues and restrictions associated with the use of cells.
Collapse
Affiliation(s)
- Ludovica Barone
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (M.G.); (B.B.)
| | - Federica Rossi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Roberto Papait
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Mario Raspanti
- Department of Medicine and Innovative Technology, University of Insubria, 21100 Varese, Italy; (M.R.); (P.A.Z.)
| | - Piero Antonio Zecca
- Department of Medicine and Innovative Technology, University of Insubria, 21100 Varese, Italy; (M.R.); (P.A.Z.)
| | - Luca Buonarrivo
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (M.G.); (B.B.)
| | - Giovanni Bernardini
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| | - Antonino Bruno
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (M.G.); (B.B.)
| | - Rosalba Gornati
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (L.B.); (F.R.); (R.P.); (L.B.); (G.B.)
| |
Collapse
|
32
|
Liu P, Wang J, Xue Y, Zou L, Tian Y, Sun R, Zhang W, Li Y, Lv L, Gao Q, Fan B. Perfusion in vivo bioreactor promotes regeneration of vascularized tissue-engineered bone. Regen Med 2023; 18:707-718. [PMID: 37589274 DOI: 10.2217/rme-2023-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Aim: This study improved the in vivo bioreactor (IVB) for bone regeneration by enhancing stem cell survival and promoting vascularized tissue-engineered bone. Methods: 12 New Zealand rabbits received β-TCP scaffolds with rabbit bone mesenchymal stem cells (BMSCs) implanted. Perfusion IVB with a perfusion electronic pump was compared with the control group using micro-CT, Microfil perfusion, histological staining and RT-PCR for gene expression. Results: Perfusion IVB demonstrated good biocompatibility, increased neoplastic bone tissue, neovascularization and upregulated osteogenic and angiogenesis-related genes in rabbits (p < 0.05). Conclusion: Perfusion IVB holds promise for bone regeneration and tissue engineering in orthopedics and maxillofacial surgery.
Collapse
Affiliation(s)
- Peng Liu
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu Province, 730050, China
| | - Jian Wang
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Yun Xue
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Lei Zou
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Yongzheng Tian
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu Province, 730050, China
| | - Ruilong Sun
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu Province, 730050, China
| | - Wenhua Zhang
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Yunfei Li
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Lijun Lv
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Qiuming Gao
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| | - Bo Fan
- Orthopedic Centre, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, 730050, China
| |
Collapse
|
33
|
Xiang H, Xu S, Zhang W, Xue X, Li Y, Lv Y, Chen J, Miao X. Dissolving microneedles for alopecia treatment. Colloids Surf B Biointerfaces 2023; 229:113475. [PMID: 37536169 DOI: 10.1016/j.colsurfb.2023.113475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023]
Abstract
Alopecia is a treatable benign disease, however, approximately 15-30% of women and 50% of men suffer from alopecia, which greatly affects patient's self-esteem and quality of life. Currently, commercial products for alopecia treatment include topical minoxidil solution, oral finasteride tablets and oral baricitinib tablets. However, the barrier of stratum corneum, systemic adverse effects and poor cure rate limit the application of commercial products. Therefore, researchers investigated the mechanism of alopecia, and developed new drugs that could target lactate dehydrogenase-related pathways, remove excessive reactive oxygen in hair follicles, and reduce the escape of hair follicle stem cells, thus injecting new strength into the treatment of alopecia. Moreover, starting from improving drug stratum corneum penetration and reducing side effects, researchers have developed hair loss treatment strategies based on dissolved microneedles (MNs), such as drug powders/microparticles, nanoparticles, biomimetic cell membranes, phototherapy and magnetically responsive soluble microneedles, which show exciting alopecia treatment effects. However, there are still some challenges in the practical application of the current alopecia treatment strategy with soluble microneedles, and further studies are needed to accelerate its clinical translation.
Collapse
Affiliation(s)
- Hong Xiang
- Marine College, Shandong University, Weihai 264209, China
| | - Sai Xu
- Marine College, Shandong University, Weihai 264209, China
| | - Weiwei Zhang
- Drug Research and Development Center, Shandong Drug and Food Vocational College, Weihai 264209, China
| | - Xinyue Xue
- Marine College, Shandong University, Weihai 264209, China
| | - Yixuan Li
- Marine College, Shandong University, Weihai 264209, China
| | - Yanyu Lv
- Drug Research and Development Center, Shandong Drug and Food Vocational College, Weihai 264209, China
| | - Jing Chen
- Marine College, Shandong University, Weihai 264209, China
| | - Xiaoqing Miao
- Marine College, Shandong University, Weihai 264209, China.
| |
Collapse
|
34
|
Augustine R, Camci-Unal G. Scaffolds with high oxygen content support osteogenic cell survival under hypoxia. Biomater Sci 2023; 11:5560-5575. [PMID: 37401619 PMCID: PMC10533211 DOI: 10.1039/d3bm00650f] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Regeneration of large bone defects is a significant clinical challenge with variable success, but tissue engineering strategies are promising for rapid and effective bone regeneration. Maintaining an adequate oxygen level within implanted scaffolds is a major obstacle in bone tissue engineering. We developed a new oxygen-generating scaffold by electrospinning polycaprolactone with calcium peroxide (CaO2) nanocuboids (CPNCs) and characterized the physical, chemical, and biological properties of the resulting composites. Our scaffolds are highly porous and composed of submicron fibers that include CPNC as confirmed with XRD and FTIR analyses. Scaffolds containing CPNC provided controlled oxygen release for 14-days and supported cell proliferation while protecting preosteoblasts from hypoxia-induced cell death. Oxygen-generating scaffolds also facilitated bone mimetic defect contraction in vitro. The results suggest that our approach can be used to develop tissue-engineered products which target bone defects.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
35
|
Vach Agocsova S, Culenova M, Birova I, Omanikova L, Moncmanova B, Danisovic L, Ziaran S, Bakos D, Alexy P. Resorbable Biomaterials Used for 3D Scaffolds in Tissue Engineering: A Review. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4267. [PMID: 37374451 PMCID: PMC10301242 DOI: 10.3390/ma16124267] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/28/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023]
Abstract
This article provides a thorough overview of the available resorbable biomaterials appropriate for producing replacements for damaged tissues. In addition, their various properties and application possibilities are discussed as well. Biomaterials are fundamental components in tissue engineering (TE) of scaffolds and play a critical role. They need to exhibit biocompatibility, bioactivity, biodegradability, and non-toxicity, to ensure their ability to function effectively with an appropriate host response. With ongoing research and advancements in biomaterials for medical implants, the objective of this review is to explore recently developed implantable scaffold materials for various tissues. The categorization of biomaterials in this paper includes fossil-based materials (e.g., PCL, PVA, PU, PEG, and PPF), natural or bio-based materials (e.g., HA, PLA, PHB, PHBV, chitosan, fibrin, collagen, starch, and hydrogels), and hybrid biomaterials (e.g., PCL/PLA, PCL/PEG, PLA/PEG, PLA/PHB PCL/collagen, PCL/chitosan, PCL/starch, and PLA/bioceramics). The application of these biomaterials in both hard and soft TE is considered, with a particular focus on their physicochemical, mechanical, and biological properties. Furthermore, the interactions between scaffolds and the host immune system in the context of scaffold-driven tissue regeneration are discussed. Additionally, the article briefly mentions the concept of in situ TE, which leverages the self-renewal capacities of affected tissues and highlights the crucial role played by biopolymer-based scaffolds in this strategy.
Collapse
Affiliation(s)
- Sara Vach Agocsova
- Institute of Natural and Synthetic Polymers, Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37 Bratislava, Slovakia; (S.V.A.); (B.M.); (D.B.); (P.A.)
| | - Martina Culenova
- Panara a.s., Krskanska 21, 949 05 Nitra, Slovakia; (I.B.); (L.O.)
| | - Ivana Birova
- Panara a.s., Krskanska 21, 949 05 Nitra, Slovakia; (I.B.); (L.O.)
| | - Leona Omanikova
- Panara a.s., Krskanska 21, 949 05 Nitra, Slovakia; (I.B.); (L.O.)
| | - Barbora Moncmanova
- Institute of Natural and Synthetic Polymers, Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37 Bratislava, Slovakia; (S.V.A.); (B.M.); (D.B.); (P.A.)
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (L.D.); (S.Z.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Stanislav Ziaran
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (L.D.); (S.Z.)
- Department of Urology, Faculty of Medicine, Comenius University, Limbova 5, 833 05 Bratislava, Slovakia
| | - Dusan Bakos
- Institute of Natural and Synthetic Polymers, Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37 Bratislava, Slovakia; (S.V.A.); (B.M.); (D.B.); (P.A.)
- Panara a.s., Krskanska 21, 949 05 Nitra, Slovakia; (I.B.); (L.O.)
| | - Pavol Alexy
- Institute of Natural and Synthetic Polymers, Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37 Bratislava, Slovakia; (S.V.A.); (B.M.); (D.B.); (P.A.)
- Panara a.s., Krskanska 21, 949 05 Nitra, Slovakia; (I.B.); (L.O.)
| |
Collapse
|
36
|
Wang W, Gao Y, Zhang M, Li Y, Tang BZ. Neutrophil-like Biomimic AIE Nanoparticles with High-Efficiency Inflammatory Cytokine Targeting Enable Precise Photothermal Therapy and Alleviation of Inflammation. ACS NANO 2023; 17:7394-7405. [PMID: 37009988 DOI: 10.1021/acsnano.2c11762] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Although photothermal therapy (PTT) has thrived as a promising treatment for drug-resistant bacterial infections by avoiding the abuse of antibiotics, the remaining challenges that limit the treatment efficiency are the poor targeting properties of infected lesions and low penetration to the cell membrane of Gram-negative bacteria. Herein, we developed a biomimetic neutrophil-like aggregation-induced emission (AIE) nanorobot (CM@AIE NPs) for precise inflammatory site homing and efficient PTT effects. Due to their surface-loaded neutrophil membranes, CM@AIE NPs can mimic the source cell and thus interact with immunomodulatory molecules that would otherwise target endogenous neutrophils. Coupled with the secondary near-infrared region absorption and excellent photothermal properties of AIE luminogens (AIEgens), precise localization, and treatment in inflammatory sites can be achieved, thereby minimizing damage to surrounding normal tissues. Moreover, CM@AIE NP-mediated PTT was stimulated in vivo by a 980 nm laser irradiation, which contributed to the extent of the therapeutic depth and limited the damage to skin tissues. The good biocompatibility and excellent in vitro and in vivo antibacterial effects prove that CM@AIE NPs can provide a strategy for broad-spectrum antibacterial applications.
Collapse
Affiliation(s)
- Wentao Wang
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Yumeng Gao
- Jiangsu Collaborative Innovation Center for Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ming Zhang
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Yuanyuan Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| |
Collapse
|
37
|
Konoe R, Morizane R. Strategies for Improving Vascularization in Kidney Organoids: A Review of Current Trends. BIOLOGY 2023; 12:503. [PMID: 37106704 PMCID: PMC10135596 DOI: 10.3390/biology12040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023]
Abstract
Kidney organoids possess the potential to revolutionize the treatment of renal diseases. However, their growth and maturation are impeded by insufficient growth of blood vessels. Through a PubMed search, we have identified 34 studies that attempted to address this challenge. Researchers are exploring various approaches including animal transplantation, organ-on-chips, and extracellular matrices (ECMs). The most prevalent method to promote the maturation and vascularization of organoids involves transplanting them into animals for in vivo culture, creating an optimal environment for organoid growth and the development of a chimeric vessel network between the host and organoids. Organ-on-chip technology permits the in vitro culture of organoids, enabling researchers to manipulate the microenvironment and investigate the key factors that influence organoid development. Lastly, ECMs have been discovered to aid the formation of blood vessels during organoid differentiation. ECMs from animal tissue have been particularly successful, although the underlying mechanisms require further research. Future research building upon these recent studies may enable the generation of functional kidney tissues for replacement therapies.
Collapse
Affiliation(s)
| | - Ryuji Morizane
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
38
|
Wang Z, Xu H, Yang H, Zhang Y, Wang X, Wang P, Xu Z, Lv D, Rong Y, Dong Y, Tang B, Hu Z, Deng W, Zhu J. Single-stage transplantation combined with epidermal stem cells promotes the survival of tissue-engineered skin by inducing early angiogenesis. Stem Cell Res Ther 2023; 14:51. [PMID: 36959609 PMCID: PMC10035248 DOI: 10.1186/s13287-023-03281-z] [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: 08/13/2022] [Accepted: 03/13/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND The composite transplantation of a split-thickness skin graft (STSG) combined with an acellular dermal matrix (ADM) is a promising repair method for full-thickness skin defects. Due to delayed vascularization of the ADM, no currently available engineered skin tissue is able to permanently cover full-thickness skin defects via a single-stage procedure. Epidermal stem cells (EpSCs) have been found to promote angiogenesis in the wound bed. Whether EpSCs can induce early angiogenesis of dermal substitutes and promote the survival of single-stage tissue-engineered skin transplantation needs to be further studied. METHODS In vitro, rat vascular endothelial cells (RVECs) were treated with the supernatant of EpSCs cultured in ADM and stimulated for 48 h. RVECs were analysed by RNA sequencing and tube formation assays. For the in vivo experiment, 75 rats were randomly divided into five groups: ADM, ADM + EpSCs (AE), STSG, ADM + STSG (AS), and ADM + STSG + EpSCs (ASE) groups. The quality of wound healing was estimated by general observation and H&E and Masson staining. The blood perfusion volume was evaluated using the LDPI system, and the expression of vascular markers was determined by immunohistochemistry (IHC). RESULTS The active substances secreted by EpSCs cultured in ADM promoted angiogenesis, as shown by tube formation experiments and RNA-seq. EpSCs promoted epithelialization of the ADM and vascularization of the ADM implant. The ASE group showed significantly increased skin graft survival, reduced skin contraction, and an improved cosmetic appearance compared with the AS group and the STSG control group. CONCLUSIONS In summary, our findings suggest that EpSCs promote the formation of new blood vessels in dermal substitutes and support one-step transplantation of tissue-engineered skin, and thereby provide new ideas for clinical application.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hailin Xu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hao Yang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yi Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoyan Wang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peng Wang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhongye Xu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dongming Lv
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanchao Rong
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunxian Dong
- Department of Plastic Surgery, Guangdong Second Provincial General Hospital, Southern Medical University, Guangzhou, China
| | - Bing Tang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhicheng Hu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Wuguo Deng
- Collaborative Innovation Center of Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Jiayuan Zhu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
39
|
Masson-Meyers DS, Tabatabaei F, Steinhaus L, Toth JM, Tayebi L. Development of fibroblast/endothelial cell-seeded collagen scaffolds for in vitro prevascularization. J Biomed Mater Res B Appl Biomater 2023; 111:633-645. [PMID: 36262080 PMCID: PMC10585651 DOI: 10.1002/jbm.b.35182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2023]
Abstract
The development of vascularized scaffolds remains one of the major challenges in tissue engineering, and co-culturing with endothelial cells is known as one of the possible approaches for this purpose. In this approach, optimization of cell culture conditions, scaffolds, and fabrication techniques is needed to develop tissue equivalents that will enable in vitro formation of a capillary network. Prevascularized equivalents will be more physiologically comparable to the native tissues and potentially prevent insufficient vascularization after implantation. This study aimed to culture human umbilical vein endothelial cells (HUVECs), alone or in co-culture with fibroblasts, on collagen scaffolds prepared by simple fabrication approaches for in vitro prevascularization. Different concentrations and ratios of HUVECs and fibroblasts seeded on collagen gel and sponge scaffolds under several culture conditions were examined. Cell viability, scaffolds morphology, and structure were analyzed. Collagen gel scaffolds showed good cell proliferation and viability, with higher proliferation rates for cells cultured in a 2:1 (fibroblasts: HUVECs) ratio and kept in endothelial cell growth medium. However, these matrices were unable to support endothelial cell sprouting. Collagen sponges were highly porous and showed good cell viability. However, they became fragile over time in culture, and they still lack signs of vascularization. Collagen scaffolds were a good platform for cell growth and viability. However, under the experimental conditions of this study, the HUVEC/fibroblast-seeded scaffolds were not suitable platforms to generate in vitro prevascularized equivalents. Our findings will be a valuable starting point to optimize culture microenvironments and scaffolds during fabrication of prevascularized scaffolds.
Collapse
Affiliation(s)
| | | | - Lane Steinhaus
- Marquette University School of Dentistry. Milwaukee, WI 53233, USA
| | - Jeffrey M. Toth
- Marquette University School of Dentistry. Milwaukee, WI 53233, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry. Milwaukee, WI 53233, USA
| |
Collapse
|
40
|
Bari E, Di Gravina GM, Scocozza F, Perteghella S, Frongia B, Tengattini S, Segale L, Torre ML, Conti M. Silk Fibroin Bioink for 3D Printing in Tissue Regeneration: Controlled Release of MSC extracellular Vesicles. Pharmaceutics 2023; 15:pharmaceutics15020383. [PMID: 36839705 PMCID: PMC9959026 DOI: 10.3390/pharmaceutics15020383] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Sodium alginate (SA)-based hydrogels are often employed as bioink for three-dimensional (3D) scaffold bioprinting. They offer a suitable environment for cell proliferation and differentiation during tissue regeneration and also control the release of growth factors and mesenchymal stem cell secretome, which is useful for scaffold biointegration. However, such hydrogels show poor mechanical properties, fast-release kinetics, and low biological performance, hampering their successful clinical application. In this work, silk fibroin (SF), a protein with excellent biomechanical properties frequently used for controlled drug release, was blended with SA to obtain improved bioink and scaffold properties. Firstly, we produced a printable SA solution containing SF capable of the conformational change from Silk I (random coil) to Silk II (β-sheet): this transition is a fundamental condition to improve the scaffold's mechanical properties. Then, the SA-SF blends' printability and shape fidelity were demonstrated, and mechanical characterization of the printed hydrogels was performed: SF significantly increased compressive elastic modulus, while no influence on tensile response was detected. Finally, the release profile of Lyosecretome-a freeze-dried formulation of MSC-secretome containing extracellular vesicles (EV)-from scaffolds was determined: SF not only dramatically slowed the EV release rate, but also modified the kinetics and mechanism release with respect to the baseline of SA hydrogel. Overall, these results lay the foundation for the development of SA-SF bioinks with modulable mechanical and EV-release properties, and their application in 3D scaffold printing.
Collapse
Affiliation(s)
- Elia Bari
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy
- Correspondence:
| | - Giulia Maria Di Gravina
- Department of Industrial and Information Engineering, University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
- PharmaExceed s.r.l., Piazza Castello 19, 27100 Pavia, Italy
| | - Benedetta Frongia
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| | - Sara Tengattini
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Lorena Segale
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy
| | - Maria Luisa Torre
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy
- PharmaExceed s.r.l., Piazza Castello 19, 27100 Pavia, Italy
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
41
|
Shakeel A, Corridon PR. Mitigating challenges and expanding the future of vascular tissue engineering-are we there yet? Front Physiol 2023; 13:1079421. [PMID: 36685187 PMCID: PMC9846051 DOI: 10.3389/fphys.2022.1079421] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Adeeba Shakeel
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter R. Corridon
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biomedical Engineering, Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
42
|
Barreto MEV, Medeiros RP, Shearer A, Fook MVL, Montazerian M, Mauro JC. Gelatin and Bioactive Glass Composites for Tissue Engineering: A Review. J Funct Biomater 2022; 14:23. [PMID: 36662070 PMCID: PMC9861949 DOI: 10.3390/jfb14010023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Nano-/micron-sized bioactive glass (BG) particles are attractive candidates for both soft and hard tissue engineering. They can chemically bond to the host tissues, enhance new tissue formation, activate cell proliferation, stimulate the genetic expression of proteins, and trigger unique anti-bacterial, anti-inflammatory, and anti-cancer functionalities. Recently, composites based on biopolymers and BG particles have been developed with various state-of-the-art techniques for tissue engineering. Gelatin, a semi-synthetic biopolymer, has attracted the attention of researchers because it is derived from the most abundant protein in the body, viz., collagen. It is a polymer that can be dissolved in water and processed to acquire different configurations, such as hydrogels, fibers, films, and scaffolds. Searching "bioactive glass gelatin" in the tile on Scopus renders 80 highly relevant articles published in the last ~10 years, which signifies the importance of such composites. First, this review addresses the basic concepts of soft and hard tissue engineering, including the healing mechanisms and limitations ahead. Then, current knowledge on gelatin/BG composites including composition, processing and properties is summarized and discussed both for soft and hard tissue applications. This review explores physical, chemical and mechanical features and ion-release effects of such composites concerning osteogenic and angiogenic responses in vivo and in vitro. Additionally, recent developments of BG/gelatin composites using 3D/4D printing for tissue engineering are presented. Finally, the perspectives and current challenges in developing desirable composites for the regeneration of different tissues are outlined.
Collapse
Affiliation(s)
- Maria E. V. Barreto
- Northeastern Laboratory for Evaluation and Development of Biomaterials (CERTBIO), Department of Materials Engineering, Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil
| | - Rebeca P. Medeiros
- Northeastern Laboratory for Evaluation and Development of Biomaterials (CERTBIO), Department of Materials Engineering, Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil
| | - Adam Shearer
- Department of Materials Science and Engineering, The Pennsylvania State University, State College, PA 16802, USA
| | - Marcus V. L. Fook
- Northeastern Laboratory for Evaluation and Development of Biomaterials (CERTBIO), Department of Materials Engineering, Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil
| | - Maziar Montazerian
- Northeastern Laboratory for Evaluation and Development of Biomaterials (CERTBIO), Department of Materials Engineering, Federal University of Campina Grande, Campina Grande 58429-900, PB, Brazil
| | - John C. Mauro
- Department of Materials Science and Engineering, The Pennsylvania State University, State College, PA 16802, USA
| |
Collapse
|
43
|
Wang K, Liu Y, Wang H, Liu Y, Yang X, Sun S. Multi-functional nanofilms capable of angiogenesis, near-infrared-triggered anti-bacterial activity and inflammatory regulation for infected wound healing. BIOMATERIALS ADVANCES 2022; 142:213154. [PMID: 36341743 DOI: 10.1016/j.bioadv.2022.213154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/02/2022] [Accepted: 10/09/2022] [Indexed: 06/16/2023]
Abstract
Chronic infected wound healing is a critical challenge in clinical practice owing to the involvement of multiple physiological processes, including bacteria-related, inflammatory regulation and angiogenesis. Therefore, a multi-functional strategy with synergistic anti-bacterial, anti-inflammatory and pro-angiogenic effects should be developed. Owing to their biomimetic structural features and controlled delivery of active agents, electrospun nanofilms are promising biomaterials for the treatment of skin defects. In this study, we fabricated multi-functional nanofilms with pro-angiogenic, anti-bacterial and anti-inflammatory capacities. First, strontium (Sr) ions were incorporated into poly(L-lactic-co-caprolactone) (PLCL) nanofilms. Subsequently, polydopamine (PDA) and zinc oxide (ZnO) were decorated onto the surface of Sr-loaded PLCL nanofilms to prepare ZnO/PDA@PLCL@Sr nanofilms. In vitro results showed that ZnO/PDA@PLCL@Sr nanofilms were biocompatible, exhibited angiogenic activity and significantly inhibited the growth of Staphylococcus aureus and Escherichia coli upon near-infrared -light irradiation. Furthermore, ZnO/PDA@PLCL@Sr nanofilms were found to drive the transformation of macrophages into the M2 phenotype. In vivo results further validated that ZnO/PDA@PLCL@Sr nanofilms exhibited pro-angiogenic and anti-bacterial activities and regulated inflammation to accelerate wound -healing in a rat model of bacteria-infected skin defects. In conclusion, we successfully developed a multi-functional biomaterial with pro-angiogenic, anti-bacterial and anti-inflammatory capacities to treat chronic infected wounds.
Collapse
Affiliation(s)
- Kun Wang
- Department of Burns and Wound Repair, Weifang People's Hospital, Weifang 261041, China
| | - Yanqun Liu
- National Tissue Engineering Center of China, Shanghai 200241, China
| | - Hui Wang
- Department of Burns and Wound Repair, Weifang People's Hospital, Weifang 261041, China
| | - Yufang Liu
- Department of Burns and Wound Repair, Weifang People's Hospital, Weifang 261041, China
| | - Xuelin Yang
- Department of Burns and Wound Repair, Weifang People's Hospital, Weifang 261041, China
| | - Shudong Sun
- Department of Burns and Wound Repair, Weifang People's Hospital, Weifang 261041, China.
| |
Collapse
|
44
|
Redmond J, McCarthy HO, Buchanan P, Levingstone TJ, Dunne NJ. Development and characterisation of 3D collagen-gelatin based scaffolds for breast cancer research. BIOMATERIALS ADVANCES 2022; 142:213157. [PMID: 36279748 DOI: 10.1016/j.bioadv.2022.213157] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/20/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
While 2D culture presents a useful tool for cancer research, it fails to replicate the tumor microenvironment as it lacks proper three-dimensional cell-cell/cell-matrix interactions, often resulting in exaggerated responses to therapeutic agents. 3D models that aim to overcome the issues associated with 2D culture research offer a new frontier for cancer research with cell growth, morphology and genetic properties that more closely match in vivo cancers. Herein, we aim to develop a collagen-based scaffold that supports the attachment and proliferation of breast cancer (BC) cells as a 3D culture model. Scaffolds were produced on a repeatable basis using a freeze-drying procedure. The constructs were highly porous (>99%) with homogenous pore sizes (150-300 μm) and an interconnected structure. The application of 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDAC) crosslinking resulted in scaffolds with elastic moduli in the range of 1-2 kPa, mimicking cancerous breast tissue stiffness. Furthermore, the incorporation of gelatin into the scaffolds enabled the porosity, pore size and mechanical properties to be tailored, resulting in scaffolds with stiffness values that accurately replicate the stiffness of human BC extracellular matrix (ECM) (1.3-1.7 kPa). Scaffolds displayed high in vitro stability with 90% of mass remaining after 14 days of culture. The scaffolds were shown to be highly biocompatible, and capable of supporting the attachment, infiltration and proliferation of MCF7 breast cancer (BC) cells over +14 days. These results confirm the suitability of these scaffolds as culture models for BC cells. These collagen-based scaffolds offer significant potential for the exploration of aspects of BC, such as gene expression profiles and patterns, and for the assessment of the efficacy of therapeutic agents in treating BC.
Collapse
Affiliation(s)
- John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, Collins Avenue, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland.
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom; School of Chemical Sciences, Dublin City University, Collins Avenue, Dublin 9, Ireland
| | - Paul Buchanan
- School of Nursing and Human Science, Dublin City University, Collins Avenue, Dublin, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Collins Avenue, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Biodesign Europe, Dublin City University, Dublin 9, Ireland.
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Collins Avenue, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland; School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Biodesign Europe, Dublin City University, Dublin 9, Ireland.
| |
Collapse
|
45
|
Khanna A, Oropeza BP, Huang NF. Engineering Spatiotemporal Control in Vascularized Tissues. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100555. [PMID: 36290523 PMCID: PMC9598830 DOI: 10.3390/bioengineering9100555] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
A major challenge in engineering scalable three-dimensional tissues is the generation of a functional and developed microvascular network for adequate perfusion of oxygen and growth factors. Current biological approaches to creating vascularized tissues include the use of vascular cells, soluble factors, and instructive biomaterials. Angiogenesis and the subsequent generation of a functional vascular bed within engineered tissues has gained attention and is actively being studied through combinations of physical and chemical signals, specifically through the presentation of topographical growth factor signals. The spatiotemporal control of angiogenic signals can generate vascular networks in large and dense engineered tissues. This review highlights the developments and studies in the spatiotemporal control of these biological approaches through the coordinated orchestration of angiogenic factors, differentiation of vascular cells, and microfabrication of complex vascular networks. Fabrication strategies to achieve spatiotemporal control of vascularization involves the incorporation or encapsulation of growth factors, topographical engineering approaches, and 3D bioprinting techniques. In this article, we highlight the vascularization of engineered tissues, with a focus on vascularized cardiac patches that are clinically scalable for myocardial repair. Finally, we discuss the present challenges for successful clinical translation of engineered tissues and biomaterials.
Collapse
Affiliation(s)
| | - Beu P. Oropeza
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Ngan F. Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
46
|
Tsiklin IL, Shabunin AV, Kolsanov AV, Volova LT. In Vivo Bone Tissue Engineering Strategies: Advances and Prospects. Polymers (Basel) 2022; 14:polym14153222. [PMID: 35956735 PMCID: PMC9370883 DOI: 10.3390/polym14153222] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/25/2022] [Accepted: 08/04/2022] [Indexed: 12/12/2022] Open
Abstract
Reconstruction of critical-sized bone defects remains a tremendous challenge for surgeons worldwide. Despite the variety of surgical techniques, current clinical strategies for bone defect repair demonstrate significant limitations and drawbacks, including donor-site morbidity, poor anatomical match, insufficient bone volume, bone graft resorption, and rejection. Bone tissue engineering (BTE) has emerged as a novel approach to guided bone tissue regeneration. BTE focuses on in vitro manipulations with seed cells, growth factors and bioactive scaffolds using bioreactors. The successful clinical translation of BTE requires overcoming a number of significant challenges. Currently, insufficient vascularization is the critical limitation for viability of the bone tissue-engineered construct. Furthermore, efficacy and safety of the scaffolds cell-seeding and exogenous growth factors administration are still controversial. The in vivo bioreactor principle (IVB) is an exceptionally promising concept for the in vivo bone tissue regeneration in a predictable patient-specific manner. This concept is based on the self-regenerative capacity of the human body, and combines flap prefabrication and axial vascularization strategies. Multiple experimental studies on in vivo BTE strategies presented in this review demonstrate the efficacy of this approach. Routine clinical application of the in vivo bioreactor principle is the future direction of BTE; however, it requires further investigation for overcoming some significant limitations.
Collapse
Affiliation(s)
- Ilya L. Tsiklin
- Biotechnology Center “Biotech”, Samara State Medical University, 443079 Samara, Russia
- City Clinical Hospital Botkin, Moscow Healthcare Department, 125284 Moscow, Russia
- Correspondence: ; Tel.: +7-903-621-81-88
| | - Aleksey V. Shabunin
- City Clinical Hospital Botkin, Moscow Healthcare Department, 125284 Moscow, Russia
| | - Alexandr V. Kolsanov
- Biotechnology Center “Biotech”, Samara State Medical University, 443079 Samara, Russia
| | - Larisa T. Volova
- Biotechnology Center “Biotech”, Samara State Medical University, 443079 Samara, Russia
| |
Collapse
|
47
|
Chen M, Jiang R, Deng N, Zhao X, Li X, Guo C. Natural polymer-based scaffolds for soft tissue repair. Front Bioeng Biotechnol 2022; 10:954699. [PMID: 35928962 PMCID: PMC9343850 DOI: 10.3389/fbioe.2022.954699] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Soft tissues such as skin, muscle, and tendon are easily damaged due to injury from physical activity and pathological lesions. For soft tissue repair and regeneration, biomaterials are often used to build scaffolds with appropriate structures and tailored functionalities that can support cell growth and new tissue formation. Among all types of scaffolds, natural polymer-based scaffolds attract much attention due to their excellent biocompatibility and tunable mechanical properties. In this comprehensive mini-review, we summarize recent progress on natural polymer-based scaffolds for soft tissue repair, focusing on clinical translations and materials design. Furthermore, the limitations and challenges, such as unsatisfied mechanical properties and unfavorable biological responses, are discussed to advance the development of novel scaffolds for soft tissue repair and regeneration toward clinical translation.
Collapse
Affiliation(s)
- Meiwen Chen
- Hangzhou Women’s Hospital, Hangzhou, Zhejiang
| | - Rui Jiang
- School of Engineering, Westlake University, Hangzhou, Zhejiang
| | - Niping Deng
- School of Engineering, Westlake University, Hangzhou, Zhejiang
| | - Xiumin Zhao
- Hangzhou Women’s Hospital, Hangzhou, Zhejiang
| | - Xiangjuan Li
- Hangzhou Women’s Hospital, Hangzhou, Zhejiang
- *Correspondence: Xiangjuan Li, ; Chengchen Guo,
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, Zhejiang
- *Correspondence: Xiangjuan Li, ; Chengchen Guo,
| |
Collapse
|
48
|
Henckes NAC, Chuang L, Bosak I, Carazzai R, Garcez T, Kuhl CP, de Oliveira FDS, Loureiro Dos Santos LA, Visioli F, Cirne-Lima EO. Tissue engineering application combining epoxidized natural rubber blend and mesenchymal stem cells in in vivo response. J Biomater Appl 2022; 37:698-711. [PMID: 35733325 DOI: 10.1177/08853282221110476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study aimed to investigate biocompatibility, integration, and tissue host response of the Poly (Lactic-co-Glycolic acid) (PLGA)/Poly (isoprene) (PI) epoxidized (PLGA/PIepox) innovative scaffold combined with adipose derived mesenchymal stem cells (ADSC). We implanted the scaffold subcutaneously on the back of 18 female rats and monitored them for up to 14 days. When compared to controls, PLGA/PIepox + ADSC demonstrated an earlier vascularization, a tendency of inflammation reduction, an adequate tissue integration, higher cell proliferation, and a tendency of expression of collagen decreasing. However, 14 days post-implantation we found similar levels of CD31, Ki67 and AE1/AE3 in PLGA/PIepox when compared to control groups. The interesting results, lead us to the assumption that PLGA/PIepox is able to provide an effective delivery system for ADSC on tissue host. This animal study assesses PLGA/PIepox + ADSC in in vivo tissue functionality and validation of use, serving as a proof of concept for future clinical translation as it presents an innovative and promising tissue engineering opportunity for the use in tissue reconstruction.
Collapse
Affiliation(s)
- Nicole Andréa Corbellini Henckes
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências da Saúde: Ginecologia e Obstetrícia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Laura Chuang
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Isadora Bosak
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Rafael Carazzai
- Laboratório de Biomateriais e Cerâmicas Avançadas, Departamento de Materiais, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tuane Garcez
- Unidade de Experimentação Animal - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Cristiana Palma Kuhl
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências da Saúde: Ginecologia e Obstetrícia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Dos Santos de Oliveira
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Luis Alberto Loureiro Dos Santos
- Laboratório de Biomateriais e Cerâmicas Avançadas, Departamento de Materiais, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Visioli
- Unidade de Patologia Experimental - Centro de Pesquisa Experimental, 37895Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Odontologia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Elizabeth Obino Cirne-Lima
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências da Saúde: Ginecologia e Obstetrícia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Patologia Clínica Veterinária, Faculdade de Veterinária, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
49
|
Fayon A, Helle D, Francius G, Vincourt JB, Regnault V, Dumas D, Menu P, El Omar R. Characterization of an Innovative Biomaterial Derived From Human Wharton’s Jelly as a New Promising Coating for Tissue Engineering Applications. Front Bioeng Biotechnol 2022; 10:884069. [PMID: 35769101 PMCID: PMC9234273 DOI: 10.3389/fbioe.2022.884069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
The extracellular matrix (ECM) offers the opportunity to create a biomaterial consisting of a microenvironment with interesting biological and biophysical properties for improving and regulating cell functions. Animal-derived ECM are the most widely used as an alternative to human tissues that are of very limited availability. However, incomplete decellularization of these tissues presents a high risk of immune rejection and disease transmission. In this study, we present an innovative method to extract human ECM derived from the Wharton’s jelly (WJ-ECMaa) of umbilical cords as a novel biomaterial to be used in tissue engineering. WJ-ECMaa was very efficiently decellularized, suggesting its possible use in allogeneic conditions. Characterization of its content allowed the identification of type I collagen as its main component. Various other matrix proteins, playing an important role in cell adhesion and proliferation, were also detected. WJ-ECMaa applied as a surface coating was analyzed by fluorescent labeling and atomic force microscopy. The results revealed a particular arrangement of collagen fibers not previously described in the literature. This biomaterial also presented better cytocompatibility compared to the conventional collagen coating. Moreover, it showed adequate hemocompatibility, allowing its use as a surface with direct contact with blood. Application of WJ-ECMaa as a coating of the luminal surface of umbilical arteries for a use in vascular tissue engineering, has improved significantly the cellularization of this surface by allowing a full and homogeneous cell coverage. Taking these results together, our novel extraction method of human ECM offers a very promising biomaterial with many potential applications in tissue engineering such as the one presented direct in vascular tissue engineering. Further characterization of the composition and functionality will help explore the ways it can be used in tissue engineering applications, especially as a scaffold or a surface coating.
Collapse
Affiliation(s)
- Adrien Fayon
- Université de Lorraine, CNRS, IMoPA, Nancy, France
| | | | - Gregory Francius
- CNRS, Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l’Environnement, Université de Lorraine, Nancy, France
| | - Jean-Baptiste Vincourt
- Université de Lorraine, CNRS, IMoPA, Nancy, France
- Université de Lorraine, CNRS, INSERM, IBSLor (UMS2008/US40), Nancy, France
| | | | | | - Patrick Menu
- Université de Lorraine, CNRS, IMoPA, Nancy, France
- *Correspondence: Patrick Menu,
| | | |
Collapse
|
50
|
Wan R, Hussain A, Behfar A, Moran SL, Zhao C. The Therapeutic Potential of Exosomes in Soft Tissue Repair and Regeneration. Int J Mol Sci 2022; 23:ijms23073869. [PMID: 35409228 PMCID: PMC8998690 DOI: 10.3390/ijms23073869] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Soft tissue defects are common following trauma and tumor extirpation. These injuries can result in poor functional recovery and lead to a diminished quality of life. The healing of skin and muscle is a complex process that, at present, leads to incomplete recovery and scarring. Regenerative medicine may offer the opportunity to improve the healing process and functional outcomes. Barriers to regenerative strategies have included cost, regulatory hurdles, and the need for cell-based therapies. In recent years, exosomes, or extracellular vesicles, have gained tremendous attention in the field of soft tissue repair and regeneration. These nanosized extracellular particles (30-140 nm) can break the cellular boundaries, as well as facilitate intracellular signal delivery in various regenerative physiologic and pathologic processes. Existing studies have established the potential of exosomes in regenerating tendons, skeletal muscles, and peripheral nerves through different mechanisms, including promoting myogenesis, increasing tenocyte differentiation and enhancing neurite outgrowth, and the proliferation of Schwann cells. These exosomes can be stored for immediate use in the operating room, and can be produced cost efficiently. In this article, we critically review the current advances of exosomes in soft tissue (tendons, skeletal muscles, and peripheral nerves) healing. Additionally, new directions for clinical applications in the future will be discussed.
Collapse
Affiliation(s)
- Rou Wan
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (R.W.); (A.H.); (S.L.M.)
| | - Arif Hussain
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (R.W.); (A.H.); (S.L.M.)
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Steven L. Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (R.W.); (A.H.); (S.L.M.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| |
Collapse
|