1
|
Xie X, Xu J, Ding D, Lin J, Han K, Wang C, Wang F, Zhao J, Wang L. Janus Membranes Patch Achieves High-Quality Tendon Repair: Inhibiting Exogenous Healing and Promoting Endogenous Healing. NANO LETTERS 2024; 24:4300-4309. [PMID: 38534038 DOI: 10.1021/acs.nanolett.4c00818] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
The imbalance between endogenous and exogenous healing is the fundamental reason for the poor tendon healing. In this study, a Janus patch was developed to promote endogenous healing and inhibit exogenous healing, leading to improved tendon repair. The upper layer of the patch is a poly(dl-lactide-co-glycolide)/polycaprolactone (PLGA/PCL) nanomembrane (PMCP-NM) modified with poly(2-methylacryloxyethyl phosphocholine) (PMPC), which created a lubricated and antifouling surface, preventing cell invasion and mechanical activation. The lower layer is a PLGA/PCL fiber membrane loaded with fibrin (Fb) (Fb-NM), serving as a temporary chemotactic scaffold to regulate the regenerative microenvironment. In vitro, the Janus patch effectively reduced 92.41% cell adhesion and 79.89% motion friction. In vivo, the patch inhibited tendon adhesion through the TGF-β/Smad signaling pathway and promoted tendon maturation. This Janus patch is expected to provide a practical basis and theoretical guidance for high-quality soft tissue repair.
Collapse
Affiliation(s)
- Xiaojing Xie
- Key Laboratory of Textile Science & Technology of Ministry of Education College of Textiles, Donghua University, Shanghai 201620, China
| | - Junjie Xu
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Danzhi Ding
- Key Laboratory of Textile Science & Technology of Ministry of Education College of Textiles, Donghua University, Shanghai 201620, China
| | - Jing Lin
- Key Laboratory of Textile Science & Technology of Ministry of Education College of Textiles, Donghua University, Shanghai 201620, China
| | - Kang Han
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Chaorong Wang
- Shanghai Frontier Science Research Center for Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Fujun Wang
- Shanghai Frontier Science Research Center for Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education College of Textiles, Donghua University, Shanghai 201620, China
- Shanghai Frontier Science Research Center for Advanced Textiles, College of Textiles, Donghua University, Shanghai 201620, China
| |
Collapse
|
2
|
Vanderslice EJ, Golding SGH, Jacot JG. Vascularization of PEGylated fibrin hydrogels increases the proliferation of human iPSC-cardiomyocytes. J Biomed Mater Res A 2024; 112:625-634. [PMID: 38155509 PMCID: PMC10922460 DOI: 10.1002/jbm.a.37662] [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: 06/14/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/30/2023]
Abstract
Studies have long sought to develop engineered heart tissue for the surgical correction of structural heart defects, as well as other applications and vascularization of this tissue has presented a challenge. Recent studies suggest that vascular cells and a vascular network may have regenerative effects on implanted cardiomyocytes (CM) and nearby heart tissue separate from perfusion of oxygen and nutrients. The goal of this study was to test whether vascular cells or a formed vascular network in a fibrin-based hydrogel would alter the proliferation of human iPSC-derived CM. First, vascular network formation in a slowly degrading PEGylated fibrin hydrogel was optimized by altering the cell ratio of human umbilical vein endothelial cells to human dermal fibroblasts, the inclusion of growth factors, and the total cell concentration. An endothelial to fibroblast ratio of 5:1 and a total cell concentration of 1.1 × 106 cells/mL without additional growth factors generated robust vascular networks while minimizing the number of cells required. Using this optimized system, human iPSC-derived CM were cultured on hydrogels without vascular cells, hydrogels with unorganized encapsulated vascular cells, or hydrogels with encapsulated vascular cells organized into networks for 7 days. CM proliferation and gene expression were assayed following 7 days of culture on the hydrogels. The presence of vascular cells in the hydrogel, whether unorganized or in vascular networks, significantly increased CM proliferation compared to an acellular hydrogel. Hydrogels with unorganized vascular cells resulted in lower CM maturity evidenced by decreased expression of cardiac troponin t (TNNT2), myosin light chain 7, and phospholamban compared to hydrogels without vascular cells and hydrogels with vascular networks. Altogether, this study details a robust method of forming rudimentary vascular networks in a fibrin-based hydrogel and shows that a hydrogel containing endothelial cells and fibroblasts can induce proliferation in adjacent CM, and these cells do not hinder CM gene expression when organized into a vascular network.
Collapse
Affiliation(s)
- Ethan J. Vanderslice
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA 80045
| | - Staunton G. H. Golding
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA 80045
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA 37235
| | - Jeffrey G. Jacot
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA 80045
- Department of Pediatrics, Children’s Hospital Colorado, Aurora, CO, USA 80045
| |
Collapse
|
3
|
Wang Y, Li J, Han H, Huang H, Du H, Cheng L, Ma C, Cai Y, Li G, Tao J, Cheng P. Application of locally responsive design of biomaterials based on microenvironmental changes in myocardial infarction. iScience 2023; 26:107662. [PMID: 37670787 PMCID: PMC10475519 DOI: 10.1016/j.isci.2023.107662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Morbidity and mortality caused by acute myocardial infarction (AMI) are on the rise, posing a grave threat to the health of the general population. Up to now, interventional, surgical, and pharmaceutical therapies have been the main treatment methods for AMI. Effective and timely reperfusion therapy decreases mortality, but it cannot stimulate myocardial cell regeneration or reverse ventricular remodeling. Cell therapy, gene therapy, immunotherapy, anti-inflammatory therapy, and several other techniques are utilized by researchers to improve patients' prognosis. In recent years, biomaterials for AMI therapy have become a hot spot in medical care. Biomaterials furnish a microenvironment conducive to cell growth and deliver therapeutic factors that stimulate cell regeneration and differentiation. Biomaterials adapt to the complex microenvironment and respond to changes in local physical and biochemical conditions. Therefore, environmental factors and material properties must be taken into account when designing biomaterials for the treatment of AMI. This article will review the factors that need to be fully considered in the design of biological materials.
Collapse
Affiliation(s)
- Yiren Wang
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Junlin Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hukui Han
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huihui Huang
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huan Du
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lianying Cheng
- Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Cui Ma
- Department of Mathematics, Army Medical University, Chongqing 400038, China
| | - Yongxiang Cai
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Gang Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jianhong Tao
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu 610072, China
| |
Collapse
|
4
|
Hodge JG, Decker HE, Robinson JL, Mellott AJ. Tissue-mimetic culture enhances mesenchymal stem cell secretome capacity to improve regenerative activity of keratinocytes and fibroblasts in vitro. Wound Repair Regen 2023; 31:367-383. [PMID: 36866522 DOI: 10.1111/wrr.13076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/18/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) are a heterogenous population of multipotent and highly secretory cells currently being investigated in the field of wound healing for their ability to augment tissue responses. The adaptive response of MSC populations to the rigid substrate of current 2D culture systems has been considered to result in a deterioration of regenerative 'stem-like' properties. In this study, we characterise how the improved culture of adipose-derived mesenchymal stem cells (ASCs) within a tissue-mimetic 3D hydrogel system, that is mechanically similar to native adipose tissue, enhances their regenerative capabilities. Notably, the hydrogel system contains a porous microarchitecture that permits mass transport, enabling efficient collection of secreted cellular compounds. By utilising this 3D system, ASCs retained a significantly higher expression of ASC 'stem-like' markers while demonstrating a significant reduction in senescent populations, relative to 2D. Additionally, culture of ASCs within the 3D system resulted in enhanced secretory activity with significant increases in the secretion of proteinaceous factors, antioxidants and extracellular vesicles (EVs) within the conditioned media (CM) fraction. Lastly, treatment of wound healing cells, keratinocytes (KCs) and fibroblasts (FBs), with ASC-CM from the 2D and 3D systems resulted in augmented functional regenerative activity, with ASC-CM from the 3D system significantly increasing KC and FB metabolic, proliferative and migratory activity. This study demonstrates the potential beneficial role of MSC culture within a tissue-mimetic 3D hydrogel system that more closely mimics native tissue mechanics, and subsequently how the improved phenotype augments secretory activity and potential wound healing capabilities of the MSC secretome.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Jennifer L Robinson
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
- Ronawk, LLC, Olathe, Kansas, USA
| |
Collapse
|
5
|
Shaik R, Xu J, Wang Y, Hong Y, Zhang G. Fibrin-Enriched Cardiac Extracellular Matrix Hydrogel Promotes In Vitro Angiogenesis. ACS Biomater Sci Eng 2023; 9:877-888. [PMID: 36630688 PMCID: PMC10064974 DOI: 10.1021/acsbiomaterials.2c01148] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiogenesis is essential for cardiac repair after myocardial infarction. Promoting angiogenesis has been demonstrated as an effective approach for myocardial infarction treatment. Several different strategies for inducing myocardial angiogenesis have been explored, including exogenous delivery of angiogenic genes, proteins, microRNAs, cells, and extracellular vesicles. Various types of injectable hydrogels have been investigated for cardiac tissue repair. One of the most promising injectable hydrogels in cardiac regeneration is a cardiac extracellular matrix hydrogel that is derived from decellularized porcine myocardium. It can be delivered minimally invasively via transendocardial delivery. The safety and efficacy of cardiac extracellular matrix hydrogels have been shown in small and large animal myocardial infarction models as well as clinical trials. The main mechanisms underlying the therapeutic benefits of cardiac extracellular matrix hydrogels have been elucidated and involved in the modulation of the immune response, downregulation of pathways related to heart failure progression and fibrosis, upregulation of genes important for cardiac muscle contraction, and enhancing cardiomyocyte differentiation and maturation from stem cells. However, no potent capillary network formation induced by cardiac extracellular matrix hydrogels has been reported. In this study, we tested the feasibility of incorporating a fibrin matrix into cardiac extracellular matrix hydrogels to improve the angiogenic properties of the hydrogel. Our in vitro results demonstrate that fibrin-enriched cardiac extracellular matrix hydrogels can induce robust endothelial cell tube formation from human umbilical vein endothelial cells and promote the sprouting of human mesenchymal stem cell spheroids. The obtained information from this study is very critical toward the future in vivo evaluation of fibrin-enriched cardiac extracellular matrix hydrogels in promoting myocardial angiogenesis.
Collapse
Affiliation(s)
- Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, State College, University Park, Pennsylvania 16801, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
6
|
Canceill T, Jourdan G, Kémoun P, Guissard C, Monsef YA, Bourdens M, Chaput B, Cavalie S, Casteilla L, Planat-Bénard V, Monsarrat P, Raymond-Letron I. Characterization and Safety Profile of a New Combined Advanced Therapeutic Medical Product Platelet Lysate-Based Fibrin Hydrogel for Mesenchymal Stromal Cell Local Delivery in Regenerative Medicine. Int J Mol Sci 2023; 24:ijms24032206. [PMID: 36768532 PMCID: PMC9916739 DOI: 10.3390/ijms24032206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (ASC) transplant to recover the optimal tissue structure/function relationship is a promising strategy to regenerate tissue lesions. Because filling local tissue defects by injection alone is often challenging, designing adequate cell carriers with suitable characteristics is critical for in situ ASC delivery. The aim of this study was to optimize the generation phase of a platelet-lysate-based fibrin hydrogel (PLFH) as a proper carrier for in situ ASC implantation and (1) to investigate in vitro PLFH biomechanical properties, cell viability, proliferation and migration sustainability, and (2) to comprehensively assess the local in vivo PLFH/ASC safety profile (local tolerance, ASC fate, biodistribution and toxicity). We first defined the experimental conditions to enhance physicochemical properties and microscopic features of PLFH as an adequate ASC vehicle. When ASC were mixed with PLFH, in vitro assays exhibited hydrogel supporting cell migration, viability and proliferation. In vivo local subcutaneous and subgingival PLFH/ASC administration in nude mice allowed us to generate biosafety data, including biodegradability, tolerance, ASC fate and engraftment, and the absence of biodistribution and toxicity to non-target tissues. Our data strongly suggest that this novel combined ATMP for in situ administration is safe with an efficient local ASC engraftment, supporting the further development for human clinical cell therapy.
Collapse
Affiliation(s)
- Thibault Canceill
- CIRIMAT, Université Toulouse III Paul Sabatier, CNRS UMR 5085, INPT, Faculté de Pharmacie, 35 Chemin des Maraichers, CEDEX 09, 31062 Toulouse, France
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
| | - Géraldine Jourdan
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Philippe Kémoun
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Christophe Guissard
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Yanad Abou Monsef
- LabHPEC, Histology and Pathology Department, Université de Toulouse, ENVT, CEDEX 03, 31076 Toulouse, France
| | - Marion Bourdens
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Benoit Chaput
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
- Service de Chirurgie Plastique, Reconstructrice et Esthétique, Centre Hospitalier Universitaire Rangueil, Avenue du Professeur Jean Poulhès, CEDEX 09, 31059 Toulouse, France
| | - Sandrine Cavalie
- CIRIMAT, Université Toulouse III Paul Sabatier, CNRS UMR 5085, INPT, Faculté de Pharmacie, 35 Chemin des Maraichers, CEDEX 09, 31062 Toulouse, France
| | - Louis Casteilla
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Valérie Planat-Bénard
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Paul Monsarrat
- Department of Oral Medicine and Toulouse University Hospital (CHU of Toulouse)—Toulouse Institute of Oral Medicine and Science, CEDEX 09, 31062 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
- Artificial and Natural Intelligence Toulouse Institute ANITI, 31400 Toulouse, France
- Correspondence:
| | - Isabelle Raymond-Letron
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Batiment INCERE, 4bis Avenue Hubert Curien, 31100 Toulouse, France
- LabHPEC, Histology and Pathology Department, Université de Toulouse, ENVT, CEDEX 03, 31076 Toulouse, France
| |
Collapse
|
7
|
Wang Z, Xiang L, Lin F, Tang Y, Cui W. 3D bioprinting of emulating homeostasis regulation for regenerative medicine applications. J Control Release 2023; 353:147-165. [PMID: 36423869 DOI: 10.1016/j.jconrel.2022.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022]
Abstract
Homeostasis is the most fundamental mechanism of physiological processes, occurring simultaneously as the production and outcomes of pathological procedures. Accompanied by manufacture and maturation of intricate and highly hierarchical architecture obtained from 3D bioprinting (three-dimension bioprinting), homeostasis has substantially determined the quality of printed tissues and organs. Instead of only shape imitation that has been the remarkable advances, fabrication for functionality to make artificial tissues and organs that act as real ones in vivo has been accepted as the optimized strategy in 3D bioprinting for the next several years. Herein, this review aims to provide not only an overview of 3D bioprinting, but also the main strategies used for homeostasis bioprinting. This paper briefly introduces the principles of 3D bioprinting system applied in homeostasis regulations firstly, and then summarizes the specific strategies and potential trend of homeostasis regulations using multiple types of stimuli-response biomaterials to maintain auto regulation, specifically displaying a brilliant prospect in hormone regulation of homeostasis with the most recently outbreak of vasculature fabrication. Finally, we discuss challenges and future prospects of homeostasis fabrication based on 3D bioprinting in regenerative medicine, hoping to further inspire the development of functional fabrication in 3D bioprinting.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lei Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Feng Lin
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
8
|
Soleimani M, Masoumi A, Momenaei B, Cheraqpour K, Koganti R, Chang AY, Ghassemi M, Djalilian AR. Applications of mesenchymal stem cells in ocular surface diseases: sources and routes of delivery. Expert Opin Biol Ther 2023; 23:509-525. [PMID: 36719365 PMCID: PMC10313829 DOI: 10.1080/14712598.2023.2175605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/30/2023] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) are novel, promising agents for treating ocular surface disorders. MSCs can be isolated from several tissues and delivered by local or systemic routes. They produce several trophic factors and cytokines, which affect immunomodulatory, transdifferentiating, angiogenic, and pro-survival pathways in their local microenvironment via paracrine secretion. Moreover, they exert their therapeutic effect through a contact-dependent manner. AREAS COVERED In this review, we discuss the characteristics, sources, delivery methods, and applications of MSCs in ocular surface disorders. We also explore the potential application of MSCs to inhibit senescence at the ocular surface. EXPERT OPINION Therapeutic application of MSCs in ocular surface disorders are currently under investigation. One major research area is corneal epitheliopathies, including chemical or thermal burns, limbal stem cell deficiency, neurotrophic keratopathy, and infectious keratitis. MSCs can promote corneal epithelial repair and prevent visually devastating sequelae of non-healing wounds. However, the optimal dosages and delivery routes have yet to be determined and further clinical trials are needed to address these fundamental questions.
Collapse
Affiliation(s)
- Mohammad Soleimani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Masoumi
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bita Momenaei
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kasra Cheraqpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Arthur Y Chang
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Mahmoud Ghassemi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
9
|
Ngarande E, Doubell E, Tamgue O, Mano M, Human P, Giacca M, Davies NH. Modified fibrin hydrogel for sustained delivery of RNAi lipopolyplexes in skeletal muscle. Regen Biomater 2022; 10:rbac101. [PMID: 36726610 PMCID: PMC9887344 DOI: 10.1093/rb/rbac101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/02/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
RNA interference is a promising therapeutical approach presently hindered by delivery concerns such as rapid RNA degradation and targeting of individual tissues. Injectable hydrogels are one potentially simple and direct route towards overcoming these barriers. Here we report on the utility of a combination of a mildly modified form of the clinically utilised fibrin hydrogel with Invivofectamine® 3.0, a lipid nonviral transfection vector, for local and sustained release. PEGylation of fibrin allowed for controlled release of small interfering RNA (siRNA)-lipopolyplexes for at least 10 days and greatly increased the stability of fibrin in vitro and in vivo. A 3D cell culture model and a release study showed transfection efficacy of siRNA-lipopolyplexes was retained for a minimum of 7 days. Injection in conjunction with PEGylated-fibrinogen significantly increased retention of siRNA-lipopolyplexes in mouse skeletal muscle and enhanced knockdown of myostatin mRNA that correlated with muscle growth. Thus, the increased efficacy observed here for the combination of a lipid nanoparticle, the only type of nonviral vector approved for the clinic, with fibrin, might allow for more rapid translation of injectable hydrogel-based RNA interference.
Collapse
Affiliation(s)
- Ellen Ngarande
- Cardiovascular Research Unit, Department of Surgery, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Emma Doubell
- Cardiovascular Research Unit, Department of Surgery, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | | | - Manuel Mano
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, WC2R 2LS, London, UK
| | - Paul Human
- Cardiovascular Research Unit, Department of Surgery, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Mauro Giacca
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, WC2R 2LS, London, UK
| | | |
Collapse
|
10
|
Hodge JG, Zamierowski DS, Robinson JL, Mellott AJ. Evaluating polymeric biomaterials to improve next generation wound dressing design. Biomater Res 2022; 26:50. [PMID: 36183134 PMCID: PMC9526981 DOI: 10.1186/s40824-022-00291-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 08/28/2022] [Indexed: 11/24/2022] Open
Abstract
Wound healing is a dynamic series of interconnected events with the ultimate goal of promoting neotissue formation and restoration of anatomical function. Yet, the complexity of wound healing can often result in development of complex, chronic wounds, which currently results in a significant strain and burden to our healthcare system. The advancement of new and effective wound care therapies remains a critical issue, with the current therapeutic modalities often remaining inadequate. Notably, the field of tissue engineering has grown significantly in the last several years, in part, due to the diverse properties and applications of polymeric biomaterials. The interdisciplinary cohesion of the chemical, biological, physical, and material sciences is pertinent to advancing our current understanding of biomaterials and generating new wound care modalities. However, there is still room for closing the gap between the clinical and material science realms in order to more effectively develop novel wound care therapies that aid in the treatment of complex wounds. Thus, in this review, we discuss key material science principles in the context of polymeric biomaterials, provide a clinical breadth to discuss how these properties affect wound dressing design, and the role of polymeric biomaterials in the innovation and design of the next generation of wound dressings.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.,Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - David S Zamierowski
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jennifer L Robinson
- Department of Chemical and Petroleum Engineering, University of Kansas, Mail Stop: 3051, 3901 Rainbow Blvd, Lawrence, KS, 66160, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
11
|
Lukomskyj AO, Rao N, Yan L, Pye JS, Li H, Wang B, Li JJ. Stem Cell-Based Tissue Engineering for the Treatment of Burn Wounds: A Systematic Review of Preclinical Studies. Stem Cell Rev Rep 2022; 18:1926-1955. [PMID: 35150392 PMCID: PMC9391245 DOI: 10.1007/s12015-022-10341-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
Abstract
Burn wounds are a devastating type of skin injury leading to severe impacts on both patients and the healthcare system. Current treatment methods are far from ideal, driving the need for tissue engineered solutions. Among various approaches, stem cell-based strategies are promising candidates for improving the treatment of burn wounds. A thorough search of the Embase, Medline, Scopus, and Web of Science databases was conducted to retrieve original research studies on stem cell-based tissue engineering treatments tested in preclinical models of burn wounds, published between January 2009 and June 2021. Of the 347 articles retrieved from the initial database search, 33 were eligible for inclusion in this review. The majority of studies used murine models with a xenogeneic graft, while a few used the porcine model. Thermal burn was the most commonly induced injury type, followed by surgical wound, and less commonly radiation burn. Most studies applied stem cell treatment immediately post-burn, with final endpoints ranging from 7 to 90 days. Mesenchymal stromal cells (MSCs) were the most common stem cell type used in the included studies. Stem cells from a variety of sources were used, most commonly from adipose tissue, bone marrow or umbilical cord, in conjunction with an extensive range of biomaterial scaffolds to treat the skin wounds. Overall, the studies showed favourable results of skin wound repair in animal models when stem cell-based tissue engineering treatments were applied, suggesting that such strategies hold promise as an improved therapy for burn wounds.
Collapse
Affiliation(s)
- Alissa Olga Lukomskyj
- Kolling Institute, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, 2065, Australia
| | - Nikitha Rao
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Lei Yan
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Jasmine Sarah Pye
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Haiyan Li
- Chemical and Environmental Engineering, School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia
| | - Bin Wang
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China.
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 315000, China.
| | - Jiao Jiao Li
- Kolling Institute, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, 2065, Australia.
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
12
|
Kim D, Hwang KS, Seo EU, Seo S, Lee BC, Choi N, Choi J, Kim HN. Vascularized Lung Cancer Model for Evaluating the Promoted Transport of Anticancer Drugs and Immune Cells in an Engineered Tumor Microenvironment. Adv Healthc Mater 2022; 11:e2102581. [PMID: 35286780 PMCID: PMC11468795 DOI: 10.1002/adhm.202102581] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/08/2022] [Indexed: 01/09/2023]
Abstract
The tumor microenvironment (TME) is the environment around the tumor, including blood vessels, immune cells, fibroblasts, signaling molecules, and the extracellular matrix (ECM). Owing to its component interactions, the TME influences tumor growth and drug delivery in a highly complex manner. Although several vascularized cancer models are developed to mimic the TME in vitro, these models cannot comprehensively reflect blood vessel-tumor spheroid interactions. Here, a method for inducing controlled tumor angiogenesis by engineering the microenvironment is presented. The interstitial flow direction regulates the direction of capillary sprouting, showing that angiogenesis occurs in the opposite direction of flow, while the existence of lung fibroblasts affects the continuity and lumen formation of sprouted capillaries. The vascularized tumor model shows enhanced delivery of anticancer drugs and immune cells to the tumor spheroids because of the perfusable vascular networks. The possibility of capillary embolism using anticancer drug-conjugated liquid metal nanoparticles is investigated using the vascularized tumor model. This vascularized tumor platform can aid in the development of effective anticancer drugs and cancer immunotherapy.
Collapse
Affiliation(s)
- Dasom Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Kyeong Seob Hwang
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
| | - Eun U Seo
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Suyeong Seo
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Program in Nano Science and TechnologyGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul08826Republic of Korea
| | - Byung Chul Lee
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Nakwon Choi
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Jonghoon Choi
- School of Integrative EngineeringChung‐Ang UniversitySeoul06974Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
13
|
Ho TC, Chang CC, Chan HP, Chung TW, Shu CW, Chuang KP, Duh TH, Yang MH, Tyan YC. Hydrogels: Properties and Applications in Biomedicine. Molecules 2022; 27:2902. [PMID: 35566251 PMCID: PMC9104731 DOI: 10.3390/molecules27092902] [Citation(s) in RCA: 229] [Impact Index Per Article: 76.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 12/19/2022] Open
Abstract
Hydrogels are crosslinked polymer chains with three-dimensional (3D) network structures, which can absorb relatively large amounts of fluid. Because of the high water content, soft structure, and porosity of hydrogels, they closely resemble living tissues. Research in recent years shows that hydrogels have been applied in various fields, such as agriculture, biomaterials, the food industry, drug delivery, tissue engineering, and regenerative medicine. Along with the underlying technology improvements of hydrogel development, hydrogels can be expected to be applied in more fields. Although not all hydrogels have good biodegradability and biocompatibility, such as synthetic hydrogels (polyvinyl alcohol, polyacrylamide, polyethylene glycol hydrogels, etc.), their biodegradability and biocompatibility can be adjusted by modification of their functional group or incorporation of natural polymers. Hence, scientists are still interested in the biomedical applications of hydrogels due to their creative adjustability for different uses. In this review, we first introduce the basic information of hydrogels, such as structure, classification, and synthesis. Then, we further describe the recent applications of hydrogels in 3D cell cultures, drug delivery, wound dressing, and tissue engineering.
Collapse
Affiliation(s)
- Tzu-Chuan Ho
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
| | - Chin-Chuan Chang
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Electrical Engineering, I-Shou University, Kaohsiung 840, Taiwan
| | - Hung-Pin Chan
- Department of Nuclear Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan;
| | - Tze-Wen Chung
- Biomedical Engineering Research and Development Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Chih-Wen Shu
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
| | - Kuo-Pin Chuang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
| | - Tsai-Hui Duh
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hui Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Center of General Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821, Taiwan
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
14
|
Cecen B, Bal-Ozturk A, Yasayan G, Alarcin E, Kocak P, Tutar R, Kozaci LD, Shin SR, Miri AK. Selection of natural biomaterials for micro-tissue and organ-on-chip models. J Biomed Mater Res A 2022; 110:1147-1165. [PMID: 35102687 PMCID: PMC10700148 DOI: 10.1002/jbm.a.37353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
The desired organ in micro-tissue models of organ-on-a-chip (OoC) devices dictates the optimum biomaterials, divided into natural and synthetic biomaterials. They can resemble biological tissues' biological functions and architectures by constructing bioactivity of macromolecules, cells, nanoparticles, and other biological agents. The inclusion of such components in OoCs allows them having biological processes, such as basic biorecognition, enzymatic cleavage, and regulated drug release. In this report, we review natural-based biomaterials that are used in OoCs and their main characteristics. We address the preparation, modification, and characterization methods of natural-based biomaterials and summarize recent reports on their applications in the design and fabrication of micro-tissue models. This article will help bioengineers select the proper biomaterials based on developing new technologies to meet clinical expectations and improve patient outcomes fusing disease modeling.
Collapse
Affiliation(s)
- Berivan Cecen
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Ayca Bal-Ozturk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey
| | - Gokcen Yasayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Polen Kocak
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Leyla Didem Kozaci
- Faculty of Medicine, Department of Medical Biochemistry, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, Massachusetts, USA
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
15
|
In Vivo Evaluation of Mechanically Processed Stromal Vascular Fraction in a Chamber Vascularized by an Arteriovenous Shunt. Pharmaceutics 2022; 14:pharmaceutics14020417. [PMID: 35214149 PMCID: PMC8880586 DOI: 10.3390/pharmaceutics14020417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a promising source for regenerative purposes. To study the in vivo fate of the mSVF, we herein used a vascularized tissue engineering chamber that insulates the target mSVF from the surrounding environment. In contrast to previous models, we propose an arteriovenous (AV) shunt between saphenous vessels in rats without a venous graft. Mechanical SVF was processed from the fat pads of male Sprague Dawley rats, mixed with a fibrin hydrogel and implanted into an inguinal tissue engineering chamber. An arteriovenous shunt was established between saphenous artery and vein. On the contralateral side, an mSVF-fibrin hydrogel mix without vascular axis served as a non-vascularized control. After two and six weeks, rats were sacrificed for further analysis. Mechanical SVF showed significant numbers of mesenchymal stromal cells. Vascularized mSVF explants gained weight over time. Perilipin and CD31 expression were significantly higher in the mSVF explants after six weeks while no difference in DAPI positive cells, collagen deposition and FABP4 expression was observed. Morphologically, no differentiated adipocytes but a dense cell-rich tissue with perilipin-positive cells was found after six weeks. The phosphorylation of ERK1/2 was significantly enhanced after six weeks while Akt activation remained unaltered. Finally, mSVF explants stably expressed and released VEGF, bFGF and TGFb. Vascularized mSVF is able to proliferate and express adipocyte-specific markers. The AV shunt model is a valuable refinement of currently existing AV loop models in the rat which contributes to the fundamental 3R principles of animal research.
Collapse
|
16
|
Raghav PK, Mann Z, Ahlawat S, Mohanty S. Mesenchymal stem cell-based nanoparticles and scaffolds in regenerative medicine. Eur J Pharmacol 2021; 918:174657. [PMID: 34871557 DOI: 10.1016/j.ejphar.2021.174657] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/05/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells owing to their regenerative potential and multilineage potency. MSCs have wide-scale applications either in their native cellular form or in conjugation with specific biomaterials as nanocomposites. Majorly, these natural or synthetic biomaterials are being used in the form of metallic and non-metallic nanoparticles (NPs) to encapsulate MSCs within hydrogels like alginate or chitosan or drug cargo loading into MSCs. In contrast, nanofibers of polymer scaffolds such as polycaprolactone (PCL), poly-lactic-co-glycolic acid (PLGA), poly-L-lactic acid (PLLA), silk fibroin, collagen, chitosan, alginate, hyaluronic acid (HA), and cellulose are used to support or grow MSCs directly on it. These MSCs based nanotherapies have application in multiple domains of biomedicine including wound healing, bone and cartilage engineering, cardiac disorders, and neurological disorders. This study focused on current approaches of MSCs-based therapies and has been divided into two major sections. The first section elaborates on MSC-based nano-therapies and their plausible applications including exosome engineering and NPs encapsulation. The following section focuses on the various MSC-based scaffold approaches in tissue engineering. Conclusively, this review mainly focused on MSC-based nanocomposite's current approaches and compared their advantages and limitations for building effective regenerative medicines.
Collapse
Affiliation(s)
- Pawan Kumar Raghav
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Zoya Mann
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Swati Ahlawat
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Sujata Mohanty
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
17
|
Gibler P, Gimble J, Hamel K, Rogers E, Henderson M, Wu X, Olesky S, Frazier T. Human Adipose-Derived Stromal/Stem Cell Culture and Analysis Methods for Adipose Tissue Modeling In Vitro: A Systematic Review. Cells 2021; 10:1378. [PMID: 34204869 PMCID: PMC8227575 DOI: 10.3390/cells10061378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Human adipose-derived stromal/stem cells (hASC) are widely used for in vitro modeling of physiologically relevant human adipose tissue. These models are useful for the development of tissue constructs for soft tissue regeneration and 3-dimensional (3D) microphysiological systems (MPS) for drug discovery. In this systematic review, we report on the current state of hASC culture and assessment methods for adipose tissue engineering using 3D MPS. Our search efforts resulted in the identification of 184 independent records, of which 27 were determined to be most relevant to the goals of the present review. Our results demonstrate a lack of consensus on methods for hASC culture and assessment for the production of physiologically relevant in vitro models of human adipose tissue. Few studies have assessed the impact of different 3D culture conditions on hASC adipogenesis. Additionally, there has been a limited use of assays for characterizing the functionality of adipose tissue in vitro. Results from this study suggest the need for more standardized culture methods and further analysis on in vitro tissue functionality. These will be necessary to validate the utility of 3D MPS as an in vitro model to reduce, refine, and replace in vivo experiments in the drug discovery regulatory process.
Collapse
Affiliation(s)
- Peyton Gibler
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Jeffrey Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Katie Hamel
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Emma Rogers
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Michael Henderson
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Spencer Olesky
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
18
|
Jarrell DK, Vanderslice EJ, Lennon ML, Lyons AC, VeDepo MC, Jacot JG. Increasing salinity of fibrinogen solvent generates stable fibrin hydrogels for cell delivery or tissue engineering. PLoS One 2021; 16:e0239242. [PMID: 34010323 PMCID: PMC8133424 DOI: 10.1371/journal.pone.0239242] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/12/2021] [Indexed: 01/27/2023] Open
Abstract
Fibrin has been used clinically for wound coverings, surgical glues, and cell delivery because of its affordability, cytocompatibility, and ability to modulate angiogenesis and inflammation. However, its rapid degradation rate has limited its usefulness as a scaffold for 3D cell culture and tissue engineering. Previous studies have sought to slow the degradation rate of fibrin with the addition of proteolysis inhibitors or synthetic crosslinkers that require multiple functionalization or polymerization steps. These strategies are difficult to implement in vivo and introduce increased complexity, both of which hinder the use of fibrin in research and medicine. Previously, we demonstrated that additional crosslinking of fibrin gels using bifunctionalized poly(ethylene glycol)-n-hydroxysuccinimide (PEG-NHS) slows the degradation rate of fibrin. In this study, we aimed to further improve the longevity of these PEG-fibrin gels such that they could be used for tissue engineering in vitro or in situ without the need for proteolysis inhibitors. It is well documented that increasing the salinity of fibrin precursor solutions affects the resulting gel morphology. Here, we investigated whether this altered morphology influences the fibrin degradation rate. Increasing the final sodium chloride (NaCl) concentration from 145 mM (physiologic level) to 250 mM resulted in fine, transparent high-salt (HS) fibrin gels that degrade 2–3 times slower than coarse, opaque physiologic-salt (PS) fibrin gels both in vitro (when treated with proteases and when seeded with amniotic fluid stem cells) and in vivo (when injected subcutaneously into mice). Increased salt concentrations did not affect the viability of encapsulated cells, the ability of encapsulated endothelial cells to form rudimentary capillary networks, or the ability of the gels to maintain induced pluripotent stem cells. Finally, when implanted subcutaneously, PS gels degraded completely within one week while HS gels remained stable and maintained viability of seeded dermal fibroblasts. To our knowledge, this is the simplest method reported for the fabrication of fibrin gels with tunable degradation properties and will be useful for implementing fibrin gels in a wide range of research and clinical applications.
Collapse
Affiliation(s)
- Dillon K. Jarrell
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Ethan J. Vanderslice
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Mallory L. Lennon
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Anne C. Lyons
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Mitchell C. VeDepo
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Jeffrey G. Jacot
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Pediatrics, Children’s Hospital Colorado, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
19
|
Krull AA, Setter DO, Gendron TF, Hrstka SCL, Polzin MJ, Hart J, Dudakovic A, Madigan NN, Dietz AB, Windebank AJ, van Wijnen AJ, Staff NP. Alterations of mesenchymal stromal cells in cerebrospinal fluid: insights from transcriptomics and an ALS clinical trial. Stem Cell Res Ther 2021; 12:187. [PMID: 33736701 PMCID: PMC7977179 DOI: 10.1186/s13287-021-02241-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have been studied with increasing intensity as clinicians and researchers strive to understand the ability of MSCs to modulate disease progression and promote tissue regeneration. As MSCs are used for diverse applications, it is important to appreciate how specific physiological environments may stimulate changes that alter the phenotype of the cells. One need for neuroregenerative applications is to characterize the spectrum of MSC responses to the cerebrospinal fluid (CSF) environment after their injection into the intrathecal space. Mechanistic understanding of cellular biology in response to the CSF environment may predict the ability of MSCs to promote injury repair or provide neuroprotection in neurodegenerative diseases. Methods In this study, we characterized changes in morphology, metabolism, and gene expression occurring in human adipose-derived MSCs cultured in human (hCSF) or artificial CSF (aCSF) as well as examined relevant protein levels in the CSF of subjects treated with MSCs for amyotrophic lateral sclerosis (ALS). Results Our results demonstrated that, under intrathecal-like conditions, MSCs retained their morphology, though they became quiescent. Large-scale transcriptomic analysis of MSCs revealed a distinct gene expression profile for cells cultured in aCSF. The aCSF culture environment induced expression of genes related to angiogenesis and immunomodulation. In addition, MSCs in aCSF expressed genes encoding nutritional growth factors to expression levels at or above those of control cells. Furthermore, we observed a dose-dependent increase in growth factors and immunomodulatory cytokines in CSF from subjects with ALS treated intrathecally with autologous MSCs. Conclusions Overall, our results suggest that MSCs injected into the intrathecal space in ongoing clinical trials remain viable and may provide a therapeutic benefit to patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02241-9.
Collapse
Affiliation(s)
- Ashley A Krull
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Deborah O Setter
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sybil C L Hrstka
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Michael J Polzin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Joseph Hart
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nicolas N Madigan
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Anthony J Windebank
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nathan P Staff
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
20
|
Yang G, Mahadik B, Choi JY, Yu JR, Mollot T, Jiang B, He X, Fisher JP. Fabrication of centimeter-sized 3D constructs with patterned endothelial cells through assembly of cell-laden microbeads as a potential bone graft. Acta Biomater 2021; 121:204-213. [PMID: 33271356 DOI: 10.1016/j.actbio.2020.11.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
Modular tissue engineering is a promising biofabrication strategy to create engineered bone grafts in a bottom-up manner, in which cell-laden micro-modules are prepared as basic building blocks to assemble macroscopic tissues via different integrating mechanisms. In this study, we prepared collagen microbeads loaded with human bone marrow derived mesenchymal stem cells (BMSCs) using a microfluidic approach. The cell-laden microbeads were characterized for size change, cell activity, osteogenesis, as well as their self-assembly properties to generate centimeter-sized constructs. Moreover, using the cell-laden beads as a supporting medium, induced pluripotent stem cell-derived endothelial cells (iPSC-EC) were patterned inside bead aggregates through extrusion-based 3D printing. This fabrication approach that combines modular tissue engineering and supports 3D printing has the potential to create 3D engineered bone grafts with a pre-existing, customized vasculature.
Collapse
Affiliation(s)
- Guang Yang
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States
| | - Bhushan Mahadik
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States
| | - Ji Young Choi
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States
| | - Justine R Yu
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States; University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Trevor Mollot
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States
| | - Bin Jiang
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States
| | - John P Fisher
- Tissue Engineering & Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, United States; NIBIB/NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
21
|
Ma J, Huang C. Composition and Mechanism of Three-Dimensional Hydrogel System in Regulating Stem Cell Fate. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:498-518. [PMID: 32272868 DOI: 10.1089/ten.teb.2020.0021] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Three-dimensional (3D) hydrogel systems integrating different types of stem cells and scaffolding biomaterials have an important application in tissue engineering. The biomimetic hydrogels that pattern cell suspensions within 3D configurations of biomaterial networks allow for the transport of bioactive factors and mimic the stem cell niche in vivo, thereby supporting the proliferation and differentiation of stem cells. The composition of a 3D hydrogel system determines the physical and chemical characteristics that regulate stem cell function through a biological mechanism. Here, we discuss the natural and synthetic hydrogel compositions that have been employed in 3D scaffolding, focusing on their characteristics, fabrication, biocompatibility, and regulatory effects on stem cell proliferation and differentiation. We also discuss the regulatory mechanisms of cell-matrix interaction and cell-cell interaction in stem cell activities in various types of 3D hydrogel systems. Understanding hydrogel compositions and their cellular mechanisms can yield insights into how scaffolding biomaterials and stem cells interact and can lead to the development of novel hydrogel systems of stem cells in tissue engineering and stem cell-based regenerative medicine. Impact statement Three-dimensional hydrogel system of stem cell mimicking the stemcell niche holds significant promise in tissue engineering and regenerative medicine. Exactly how hydrogel composition regulates stem cell fate is not well understood. This review focuses on the composition of hydrogel, and how the hydrogel composition and its properties regulate the stem cell adhesion, growth, and differentiation. We propose that cell-matrix interaction and cell-cell interaction are important regulatory mechanisms in stem cell activities. Our review provides key insights into how the hydrogel composition regulates the stem cell fate, untangling the engineering of three-dimensional hydrogel systems for stem cells.
Collapse
Affiliation(s)
- Jianrui Ma
- Center for Neurobiology, Shantou University Medical College, Shantou, China
| | - Chengyang Huang
- Center for Neurobiology, Shantou University Medical College, Shantou, China
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
22
|
de Melo BA, Jodat YA, Cruz EM, Benincasa JC, Shin SR, Porcionatto MA. Strategies to use fibrinogen as bioink for 3D bioprinting fibrin-based soft and hard tissues. Acta Biomater 2020; 117:60-76. [PMID: 32949823 DOI: 10.1016/j.actbio.2020.09.024] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Fibrin gel has been widely used for engineering various types of tissues due to its biocompatible nature, biodegradability, and tunable mechanical and nanofibrous structural properties. Despite their promising regenerative capacity and extensive biocompatibility with various tissue types, fibrin-based biomaterials are often notoriously known as burdensome candidates for 3D biofabrication and bioprinting. The high viscosity of fibrin (crosslinked form) hinders proper ink extrusion, and its pre-polymer form, fibrinogen, is not capable of maintaining shape fidelity. To overcome these limitations and empower fibrinogen-based bioinks for fibrin biomimetics and regenerative applications, different strategies can be practiced. The aim of this review is to report the strategies that bring fabrication compatibility to these bioinks through mixing fibrinogen with printable biomaterials, using supporting bath supplemented with crosslinking agents, and crosslinking fibrin in situ. Moreover, the review discusses some of the recent advances in 3D bioprinting of biomimetic soft and hard tissues using fibrinogen-based bioinks, and highlights the impacts of these strategies on fibrin properties, its bioactivity, and the functionality of the consequent biomimetic tissue. Statement of Significance Due to its biocompatible nature, biodegradability, and tunable mechanical and nanofibrous structural properties, fibrin gel has been widely employed in tissue engineering and more recently, used as in 3D bioprinting. The fibrinogen's poor printable properties make it difficult to maintain the 3D shape of bioprinted constructs. Our work describes the strategies employed in tissue engineering to allow the 3D bioprinting of fibrinogen-based bioinks, such as the combination of fibrinogen with printable biomaterials, the in situ fibrin crosslinking, and the use of supporting bath supplemented with crosslinking agents. Further, this review discuss the application of 3D bioprinting technology to biofabricate fibrin-based soft and hard tissues for biomedical applications, and discuss current limitations and future of such in vitro models.
Collapse
|
23
|
Tavakoli J, Wang J, Chuah C, Tang Y. Natural-based Hydrogels: A Journey from Simple to Smart Networks for Medical Examination. Curr Med Chem 2020; 27:2704-2733. [PMID: 31418656 DOI: 10.2174/0929867326666190816125144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Natural hydrogels, due to their unique biological properties, have been used extensively for various medical and clinical examinations that are performed to investigate the signs of disease. Recently, complex-crosslinking strategies improved the mechanical properties and advanced approaches have resulted in the introduction of naturally derived hydrogels that exhibit high biocompatibility, with shape memory and self-healing characteristics. Moreover, the creation of self-assembled natural hydrogels under physiological conditions has provided the opportunity to engineer fine-tuning properties. To highlight recent studies of natural-based hydrogels and their applications for medical investigation, a critical review was undertaken using published papers from the Science Direct database. This review presents different natural-based hydrogels (natural, natural-synthetic hybrid and complex-crosslinked hydrogels), their historical evolution, and recent studies of medical examination applications. The application of natural-based hydrogels in the design and fabrication of biosensors, catheters and medical electrodes, detection of cancer, targeted delivery of imaging compounds (bioimaging) and fabrication of fluorescent bioprobes is summarised here. Without doubt, in future, more useful and practical concepts will be derived to identify natural-based hydrogels for a wide range of clinical examination applications.
Collapse
Affiliation(s)
- Javad Tavakoli
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia.,School of Biomedical Engineering, University of Technology Sydney, Ultimo, 2007 NSW, Australia
| | - Jing Wang
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia.,Key Laboratory of Advanced Textile Composite Materials of Ministry of Education, Institute of Textile Composite, School of Textile, Tianjin Polytechnic University, Tianjin 300387, China
| | - Clarence Chuah
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia
| | - Youhong Tang
- Institute of NanoScale Science and Technology, Medical Device Research Institute, College of Science and Engineering, Flinders University, South Australia 5042, Australia
| |
Collapse
|
24
|
Temporal changes guided by mesenchymal stem cells on a 3D microgel platform enhance angiogenesis in vivo at a low-cell dose. Proc Natl Acad Sci U S A 2020; 117:19033-19044. [PMID: 32709748 PMCID: PMC7430977 DOI: 10.1073/pnas.2008245117] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Therapeutic factors secreted by mesenchymal stem cells (MSCs) promote angiogenesis in vivo. However, delivery of MSCs in the absence of a cytoprotective environment offers limited efficacy due to low cell retention, poor graft survival, and the nonmaintenance of a physiologically relevant dose of growth factors at the injury site. The delivery of stem cells on an extracellular matrix (ECM)-based platform alters cell behavior, including migration, proliferation, and paracrine activity, which are essential for angiogenesis. We demonstrate the biophysical and biochemical effects of preconditioning human MSCs (hMSCs) for 96 h on a three-dimensional (3D) ECM-based microgel platform. By altering the macromolecular concentration surrounding cells in the microgels, the proangiogenic phenotype of hMSCs can be tuned in a controlled manner through cell-driven changes in extracellular stiffness and "outside-in" integrin signaling. The softest microgels were tested at a low cell dose (5 × 104 cells) in a preclinical hindlimb ischemia model showing accelerated formation of new blood vessels with a reduced inflammatory response impeding progression of tissue damage. Molecular analysis revealed that several key mediators of angiogenesis were up-regulated in the low-cell-dose microgel group, providing a mechanistic insight of pathways modulated in vivo. Our research adds to current knowledge in cell-encapsulation strategies by highlighting the importance of preconditioning or priming the capacity of biomaterials through cell-material interactions. Obtaining therapeutic efficacy at a low cell dose in the microgel platform is a promising clinical route that would aid faster tissue repair and reperfusion in "no-option" patients suffering from peripheral arterial diseases, such as critical limb ischemia (CLI).
Collapse
|
25
|
Role of biomechanics in vascularization of tissue-engineered bones. J Biomech 2020; 110:109920. [PMID: 32827778 DOI: 10.1016/j.jbiomech.2020.109920] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/23/2022]
Abstract
Biomaterial based reconstruction is still the most commonly employed method of small bone defect reconstruction. Bone tissue-engineered techniques are improving, and adjuncts such as vascularization technologies allow re-evaluation of traditional reconstructive methods for healingofcritical-sized bone defect. Slow infiltration rate of vasculogenesis after cell-seeded scaffold implantation limits the use of clinically relevant large-sized scaffolds. Hence, in vitro vascularization within the tissue-engineered bone before implantation is required to overcome the serious challenge of low cell survival rate after implantation which affects bone tissue regeneration and osseointegration. Mechanobiological interactions between cells and microvascular mechanics regulate biological processes regarding cell behavior. In addition, load-bearing scaffolds demand mechanical stability properties after vascularization to have adequate strength while implanted. With the advent of bioreactors, vascularization has been greatly improved by biomechanical regulation of stem cell differentiation through fluid-induced shear stress and synergizing osteogenic and angiogenic differentiation in multispecies coculture cells. The benefits of vascularization are clear: avoidance of mass transfer limitation and oxygen deprivation, a significant decrease in cell necrosis, and consequently bone development, regeneration and remodeling. Here, we discuss specific techniques to avoid pitfalls and optimize vascularization results of tissue-engineered bone. Cell source, scaffold modifications and bioreactor design, and technique specifics all play a critical role in this new, and rapidly growing method for bone defect reconstruction. Given the crucial importance of long-term survival of vascular network in physiological function of 3D engineered-bone constructs, greater knowledge of vascularization approaches may lead to the development of new strategies towards stabilization of formed vascular structure.
Collapse
|
26
|
Morrissette-McAlmon J, Ginn B, Somers S, Fukunishi T, Thanitcul C, Rindone A, Hibino N, Tung L, Mao HQ, Grayson W. Biomimetic Model of Contractile Cardiac Tissue with Endothelial Networks Stabilized by Adipose-Derived Stromal/Stem Cells. Sci Rep 2020; 10:8387. [PMID: 32433563 PMCID: PMC7239907 DOI: 10.1038/s41598-020-65064-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 04/23/2020] [Indexed: 11/15/2022] Open
Abstract
Cardiac tissue engineering strategies have the potential to regenerate functional myocardium following myocardial infarction. In this study, we utilized novel electrospun fibrin microfiber sheets of different stiffnesses (50.0 ± 11.2 kPa and 90.0 ± 16.4 kPa) to engineer biomimetic models of vascularized cardiac tissues. We characterized tissue assembly, electrophysiology, and contractility of neonatal rat ventricular cardiomyocytes (NRVCMs) cultured on these sheets. NRVCMs cultured on the softer substrates displayed higher conduction velocities (CVs) and improved electrophysiological properties. Human umbilical vein endothelial cells (HUVECs) formed dense networks on the sheets when co-cultured with human adipose-derived stem/stromal cells (hASCs). To achieve vascularized cardiac tissues, we tested various tri-culture protocols of NRVCM:hASC:HUVEC and found that a ratio of 1,500,000:37,500:150,000 cells/cm2 enabled the formation of robust endothelial networks while retaining statistically identical electrophysiological characteristics to NRVCM-only cultures. Tri-cultures at this ratio on 90 kPa substrates exhibited average CVs of 14 ± 0.6 cm/s, Action Potential Duration (APD)80 and APD30 of 152 ± 11 ms and 71 ± 6 ms, respectively, and maximum capture rate (MCR) of 3.9 ± 0.7 Hz. These data indicate the significant potential of generating densely packed endothelial networks together with electrically integrated cardiac cells in vitro as a physiologic 3D cardiac model.
Collapse
Affiliation(s)
- Justin Morrissette-McAlmon
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian Ginn
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD, USA
| | - Sarah Somers
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takuma Fukunishi
- Department of Surgery & Cardiac Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chanon Thanitcul
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandra Rindone
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Narutoshi Hibino
- Department of Surgery & Cardiac Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hai-Quan Mao
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD, USA.
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD, USA.
| |
Collapse
|
27
|
Huayllani MT, Sarabia-Estrada R, Restrepo DJ, Boczar D, Sisti A, Nguyen JH, Rinker BD, Moran SL, Quiñones-Hinojosa A, Forte AJ. Adipose-derived stem cells in wound healing of full-thickness skin defects: a review of the literature . J Plast Surg Hand Surg 2020; 54:263-279. [PMID: 32427016 DOI: 10.1080/2000656x.2020.1767116] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The complex process of wound healing can be delayed in circumstances when the natural niche is extremely altered. Adipose-derived stem cells (ADSC) seem to be a promising therapy for these type of wounds. We aim to describe the studies that used ADSC for wound healing after a full-thickness skin defect, the ADSC mechanisms of action, and the outcomes of the different ADSC therapies applied to date. We performed a review by querying PubMed database for studies that evaluated the use of ADSC for wound healing. The Mesh terms, adipose stem cells AND (skin injury OR wound healing) and synonyms were used for the search. Our search recorded 312 articles. A total of 30 articles met the inclusion criteria. All were experimental in nature. ADSC was applied directly (5 [16.7%]), in sheets (2 [6.7%]), scaffolds (14 [46.7%]), skin grafts (3 [10%]), skin flaps (1 [3.3%]), as microvesicles or exosomes (4 [13.3%]), with adhesives for wound closure (1 [3.3%]), and in a concentrated conditioned hypoxia-preconditioned medium (1 [3.3%]). Most of the studies reported a benefit of ADSC and improvement of wound healing with all types of ADSC therapy. ADSC applied along with extracellular matrix, stromal cell-derived factor (SDF-1) or keratinocytes, or ADSC seeded in scaffolds showed better outcomes in wound healing than ADSC alone. ADSC have shown to promote angiogenesis, fibroblast migration, and up-regulation of macrophages chemotaxis to enhance the wound healing process. Further studies should be conducted to assure the efficacy and safety of the different ADSC therapies.
Collapse
Affiliation(s)
| | | | | | - Daniel Boczar
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Andrea Sisti
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Justin H Nguyen
- Department of Transplantation Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Brian D Rinker
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Steven L Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
28
|
Shpichka AI, Konarev PV, Efremov YM, Kryukova AE, Aksenova NA, Kotova SL, Frolova AA, Kosheleva NV, Zhigalina OM, Yusupov VI, Khmelenin DN, Koroleva A, Volkov VV, Asadchikov VE, Timashev PS. Digging deeper: structural background of PEGylated fibrin gels in cell migration and lumenogenesis. RSC Adv 2020; 10:4190-4200. [PMID: 35495227 PMCID: PMC9049040 DOI: 10.1039/c9ra08169k] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/19/2019] [Indexed: 12/02/2022] Open
Abstract
Fibrin is a well-known tool in tissue engineering, but the structure of its modifications created to improve its properties remains undiscussed despite its importance, e.g. in designing biomaterials that ensure cell migration and lumenogenesis. We sought to uncover the structural aspects of PEGylated fibrin hydrogels shown to contribute to angiogenesis. The analysis of the small-angle X-ray scattering (SAXS) data and ab initio modeling revealed that the PEGylation of fibrinogen led to the formation of oligomeric species, which are larger at a higher PEG : fibrinogen molar ratio. The improvement of optical properties was provided by the decrease in aggregates' sizes and also by retaining the bound water. Compared to the native fibrin, the structure of the 5 : 1 PEGylated fibrin gel consisted of homogenously distributed flexible fibrils with a smaller space between them. Moreover, as arginylglycylaspartic acid (RGD) sites may be partly bound to PEG-NHS or masked because of the oligomerization, the number of adhesion sites may be slightly reduced that may provide the better cell migration and formation of continuous capillary-like structures.
Collapse
Affiliation(s)
- A I Shpichka
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
| | - P V Konarev
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
- National Research Center "Kurchatov Institute" Moscow Russia
| | - Yu M Efremov
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
| | - A E Kryukova
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
- National Research Center "Kurchatov Institute" Moscow Russia
| | - N A Aksenova
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
- Department of Polymers and Composites, N. N. Semenov Institute of Chemical Physics Moscow Russia
| | - S L Kotova
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
- Department of Polymers and Composites, N. N. Semenov Institute of Chemical Physics Moscow Russia
| | - A A Frolova
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
| | - N V Kosheleva
- FSBSI 'Institute of General Pathology and Pathophysiology' Moscow Russia
- Faculty of Biology, Lomonosov Moscow State University Moscow Russia
| | - O M Zhigalina
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
- Bauman Moscow State Technical University Moscow Russia
| | - V I Yusupov
- Institute of Photon Technologies, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - D N Khmelenin
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - A Koroleva
- Laser Zentrum Hannover e. V. Hannover Germany
| | - V V Volkov
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - V E Asadchikov
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - P S Timashev
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
- Department of Polymers and Composites, N. N. Semenov Institute of Chemical Physics Moscow Russia
- Institute of Photon Technologies, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| |
Collapse
|
29
|
Egorikhina MN, Aleynik DY, Rubtsova YP, Levin GY, Charykova IN, Semenycheva LL, Bugrova ML, Zakharychev EA. Hydrogel scaffolds based on blood plasma cryoprecipitate and collagen derived from various sources: Structural, mechanical and biological characteristics. Bioact Mater 2019; 4:334-345. [PMID: 31720490 PMCID: PMC6838346 DOI: 10.1016/j.bioactmat.2019.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/04/2019] [Accepted: 10/13/2019] [Indexed: 01/19/2023] Open
Abstract
At present there is a growing need for tissue engineering products, including the products of scaffold-technologies. Biopolymer hydrogel scaffolds have a number of advantages and are increasingly being used to provide means of cell transfer for therapeutic treatments and for inducing tissue regeneration. This work presents original hydrogel biopolymer scaffolds based on a blood plasma cryoprecipitate and collagen and formed under conditions of enzymatic hydrolysis. Two differently originated collagens were used for the scaffold formation. During this work the structural and mechanical characteristics of the scaffold were studied. It was found that, depending on the origin of collagen, scaffolds possess differences in their structural and mechanical characteristics. Both types of hydrogel scaffolds have good biocompatibility and provide conditions that maintain the three-dimensional growth of adipose tissue stem cells. Hence, scaffolds based on such a blood plasma cryoprecipitate and collagen have good prospects as cell carriers and can be widely used in regenerative medicine.
Collapse
Affiliation(s)
- Marfa N. Egorikhina
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Diana Ya Aleynik
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Yulia P. Rubtsova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Grigory Ya Levin
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | - Irina N. Charykova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | | | - Marina L. Bugrova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, Nizhny Novgorod, Russian Federation
| | | |
Collapse
|
30
|
Ramaswamy AK, Sides RE, Cunnane EM, Lorentz KL, Reines LM, Vorp DA, Weinbaum JS. Adipose-derived stromal cell secreted factors induce the elastogenesis cascade within 3D aortic smooth muscle cell constructs. Matrix Biol Plus 2019; 4:100014. [PMID: 33543011 PMCID: PMC7852215 DOI: 10.1016/j.mbplus.2019.100014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/19/2019] [Accepted: 08/28/2019] [Indexed: 02/07/2023] Open
Abstract
Objective Elastogenesis within the medial layer of the aortic wall involves a cascade of events orchestrated primarily by smooth muscle cells, including transcription of elastin and a cadre of elastin chaperone matricellular proteins, deposition and cross-linking of tropoelastin coacervates, and maturation of extracellular matrix fiber structures to form mechanically competent vascular tissue. Elastic fiber disruption is associated with aortic aneurysm; in aneurysmal disease a thin and weakened wall leads to a high risk of rupture if left untreated, and non-surgical treatments for small aortic aneurysms are currently limited. This study analyzed the effect of adipose-derived stromal cell secreted factors on each step of the smooth muscle cell elastogenesis cascade within a three-dimensional fibrin gel culture platform. Approach and results We demonstrate that adipose-derived stromal cell secreted factors induce an increase in smooth muscle cell transcription of tropoelastin, fibrillin-1, and chaperone proteins fibulin-5, lysyl oxidase, and lysyl oxidase-like 1, formation of extracellular elastic fibers, insoluble elastin and collagen protein fractions in dynamically-active 30-day constructs, and a mechanically competent matrix after 30 days in culture. Conclusion Our results reveal a potential avenue for an elastin-targeted small aortic aneurysm therapeutic, acting as a supplement to the currently employed passive monitoring strategy. Additionally, the elastogenesis analysis workflow explored here could guide future mechanistic studies of elastin formation, which in turn could lead to new non-surgical treatment strategies. Stromal cells stimulate smooth muscle cells (SMC) using paracrine signals. Stimulated SMC make RNA for both elastin and associated proteins. After protein synthesis, new elastic fibers form that contain insoluble elastin. Stromal cell products could promote elastin production in vivo.
Collapse
Key Words
- AA, aortic aneurysm
- ACA, epsilon-amino caproic acid
- ASC, adipose-derived stromal cell
- ASC-SF, ASC secreted factors
- Aneurysm
- Aorta
- ECM, extracellular matrix
- Elastin
- Extracellular matrix
- FBS, fetal bovine serum
- LOX, lysyl oxidase
- LOXL-1, LOX-like 1
- LTBP, latent TGF-β binding protein
- NCM, non-conditioned media
- NT, no treatment
- PBS, phosphate buffered saline
- RT, reverse transcriptase
- SMC, smooth muscle cell
- TGF-β, transforming growth factor-β
- Vascular regeneration
- qPCR, quantitative polymerase chain reaction
Collapse
Affiliation(s)
- Aneesh K. Ramaswamy
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Rachel E. Sides
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Eoghan M. Cunnane
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine L. Lorentz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Leila M. Reines
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - David A. Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Justin S. Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Corresponding author at: Department of Bioengineering, University of Pittsburgh, Center for Bioengineering, Suite 300, 300 Technology Drive, Pittsburgh, PA 15261, United States of America.
| |
Collapse
|
31
|
Dhada KS, Hernandez DS, Suggs LJ. In Vivo Photoacoustic Tracking of Mesenchymal Stem Cell Viability. ACS NANO 2019; 13:7791-7799. [PMID: 31250647 PMCID: PMC7155740 DOI: 10.1021/acsnano.9b01802] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Adult stem cell therapy has demonstrated improved outcomes for treating cardiovascular diseases in preclinical trials. The development of imaging tools may increase our understanding of the mechanisms of stem cell therapy, and a variety of imaging tools have been developed to image transplanted stem cells in vivo; however, they lack the ability to interrogate stem cell function longitudinally. Here, we report the use of a nanoparticle-based contrast agent that can track stem cell viability using photoacoustic imaging. The contrast agent consists of inert gold nanorods coated with IR775c, a reactive oxygen species (ROS) sensitive near-infrared dye. Upon cell death, stem cells produce ROS to degrade the cell. Using this feature of stem cells, the viability can be measured by comparing the IR775c signal to the ROS insensitive gold nanorod signal, which can also be used to track stem cell location. The nanoprobe was successfully loaded into mesenchymal stem cells (MSCs), and then, MSCs were transplanted into the lower limb of a mouse and imaged using combined ultrasound and photoacoustic imaging. MSC viability was assessed using the nanoprobe and displayed significant cell death within 24 h and an estimated 5% viability after 10 days. This nanoparticle system allows for longitudinal tracking of MSC viability in vivo with high spatial and temporal resolution which other imaging modalities currently cannot achieve.
Collapse
Affiliation(s)
- Kabir S. Dhada
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Derek S. Hernandez
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | | |
Collapse
|
32
|
Shukla S, Mittal SK, Foulsham W, Elbasiony E, Singhania D, Sahu SK, Chauhan SK. Therapeutic efficacy of different routes of mesenchymal stem cell administration in corneal injury. Ocul Surf 2019; 17:729-736. [PMID: 31279065 DOI: 10.1016/j.jtos.2019.07.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Corneal injuries are associated with significant impairment in vision. Mesenchymal stem cells (MSCs) have been shown to limit inflammation and promote tissue repair at the ocular surface. Here, we evaluate the efficacies of different modes of MSC delivery (topical, subconjunctival, intraperitoneal [IP] and intravenous [IV]) to promote tissue repair and restore corneal transparency in a murine model of corneal injury. METHODS MSCs were purified from the bone marrow of C57BL/6 mice and expanded using plastic adherence in vitro. Corneal injury was created using an Algerbrush, and 0.5 × 106 MSCs/mouse were administered via topical, subconjunctival, IP or IV routes. Qdot-labeled MSCs were employed to determine the effect of route of administration on corneal and conjunctival MSC frequencies. Corneal opacity scores were calculated using ImageJ. Expression of inflammatory cytokines was quantified by qPCR, and infiltration of CD45+ cells was evaluated by flow cytometry. RESULTS Subconjunctival or IV administration results in increased frequencies of MSCs in ocular surface tissues following corneal injury, relative to topical or intraperitoneal delivery. Subconjunctival or IV administration reduces: (i) corneal opacity, (ii) tissue fibrosis as quantified by α-Sma expression, (iii) the expression of inflammatory cytokines (Il-1β and Tnf-α) and (iv) CD45+ inflammatory cell infiltration relative to untreated injured control animals. Administration via subconjunctival or IV routes was observed to accelerate corneal repair by restoring tissue architecture and epithelial integrity. CONCLUSIONS Our data suggest that subconjunctival or IV delivery of MSCs has superior therapeutic efficacy compared to topical or IP delivery following corneal injury.
Collapse
Affiliation(s)
- Sachin Shukla
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Center for Ocular Regeneration, L. V. Prasad Eye Institute, Hyderabad, India
| | - Sharad K Mittal
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Institute of Ophthalmology, University College London, London, UK
| | - Elsayed Elbasiony
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Disha Singhania
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Srikant K Sahu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; L.V. Prasad Eye Institute, Bhubaneswar, Odisha, India.
| | - Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Natesan S, Stone R, Coronado RE, Wrice NL, Kowalczewski AC, Zamora DO, Christy RJ. PEGylated Platelet-Free Blood Plasma-Based Hydrogels for Full-Thickness Wound Regeneration. Adv Wound Care (New Rochelle) 2019; 8:323-340. [PMID: 31737420 DOI: 10.1089/wound.2018.0844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/23/2018] [Indexed: 02/03/2023] Open
Abstract
Objective: To develop a cost-effective and clinically usable therapy to treat full-thickness skin injuries. We accomplished this by preparing a viscoelastic hydrogel using polyethylene glycol (PEG)-modified platelet-free plasma (PEGylated PFP) combined with human adipose-derived stem cells (ASCs). Approach: PEGylated PFP hydrogels were prepared by polymerizing the liquid mixture of PEG and PFP±ASCs and gelled either by adding calcium chloride (CaCl2) or thrombin. Rheological and in vitro studies were performed to assess viscoelasticity and the ability of hydrogels to direct ASCs toward a vasculogenic phenotype, respectively. Finally, a pilot study evaluated the efficacy of hydrogels±ASCs using an athymic rat full-thickness skin wound model. Results: Hydrogels prepared within the range of 11 to 27 mM for CaCl2 or 5 to 12.5 U/mL for thrombin exhibited a storage modulus of ∼62 to 87 Pa and ∼47 to 92 Pa, respectively. The PEGylated PFP hydrogels directed ASCs to form network-like structures resembling vasculature, with a fourfold increase in perivascular specific genes that were confirmed by immunofluorescent staining. Hydrogels combined with ASCs exhibited an increase in blood vessel density when applied to excisional rat wounds compared with those treated with hydrogels (110.3 vs. 95.6 BV/mm2; p < 0.05). Furthermore, ASCs were identified in the perivascular region associated with newly forming blood vessels. Innovation: This study demonstrates that PFP modified with PEG along with ASCs can be used to prepare cost-effective stable hydrogels, at the bed-side, to treat extensive skin wounds. Conclusion: These results indicate that PEGylated plasma-based hydrogels combined with ASCs may be a potential regenerative therapy for full-thickness skin wounds.
Collapse
Affiliation(s)
- Shanmugasundaram Natesan
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Randolph Stone
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | | | - Nicole L. Wrice
- Ocular Trauma & Vision Restoration, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Andrew C. Kowalczewski
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - David O. Zamora
- Ocular Trauma & Vision Restoration, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Robert J. Christy
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|
34
|
Bandara N, Gurusinghe S, Kong A, Mitchell G, Wang LX, Lim SY, Strappe P. Generation of a nitric oxide signaling pathway in mesenchymal stem cells promotes endothelial lineage commitment. J Cell Physiol 2019; 234:20392-20407. [PMID: 30997675 DOI: 10.1002/jcp.28640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
Enhancing differentiation of mesenchymal stem cells (MSCs) to endothelial cells may improve their ability to vascularize tissue and promote wound healing. This study describes a novel role for nitric oxide (NO) in reprogramming MSCs towards an endothelial lineage and highlights the role of Wnt signaling and epigenetic modification by NO. Rat MSCs were transduced with lentiviral vectors expressing endothelial nitric oxide synthase (pLV-eNOS) and a mutated caveolin gene (pLV-CAV-1F92A ) to enhance NO generation resulting in increased in vitro capillary tubule formation and endothelial marker gene expression. An exogenous source of NO could also stimulate CD31 expression in MSCs. NO was associated with an arterial-specific endothelial gene expression profile of Notch1, Dll4, and Hey2 and significantly reduced expression of venous markers. Wnt signaling associated with NO was evident through increased gene expression of Wnt3a and β-catenin protein, and expression of the endothelial marker Pecam-1 could be significantly reduced by treatment with the Wnt signaling inhibitor Dkk-1. The role of NO as an epigenetic modifier was evident with reduced gene expression of the methyltransferase, DNMT1, and bisulfite sequencing of the endothelial Flt1 promoter region in NO-producing MSCs showed significant demethylation compared to control cells. Finally, subcutaneous implantation of NO-producing MSCs seeded in a biomaterial scaffold (NovoSorb®) resulted in survival of transplanted cells and the formation of blood vessels. In summary, this study describes, NO as a potent endothelial programming factor which acts as an epigenetic modifier in MSCs and may provide a novel platform for vascular regenerative therapy.
Collapse
Affiliation(s)
- Nadeeka Bandara
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Saliya Gurusinghe
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia.,School of Agricultural and Wine Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Anne Kong
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Geraldine Mitchell
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - Le-Xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Padraig Strappe
- School of Health, Medicine and Applied Sciences, Central Queensland University, Rockhampton, Queensland, Australia
| |
Collapse
|
35
|
Piard C, Baker H, Kamalitdinov T, Fisher J. Bioprinted osteon-like scaffolds enhance in vivo neovascularization. Biofabrication 2019; 11:025013. [PMID: 30769337 PMCID: PMC7195919 DOI: 10.1088/1758-5090/ab078a] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone tissue engineers are facing a daunting challenge when attempting to fabricate bigger constructs intended for use in the treatment of large bone defects, which is the vascularization of the graft. Cell-based approaches and, in particular, the use of in vitro coculture of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) has been one of the most explored options. We present in this paper an alternative method to mimic the spatial pattern of HUVECs and hMSCs found in native osteons based on the use of extrusion-based 3D bioprinting (3DP). We developed a 3DP biphasic osteon-like scaffold, containing two separate osteogenic and vasculogenic cell populations encapsulated in a fibrin bioink in order to improve neovascularization. To this end, we optimized the fibrin bioink to improve the resolution of printed strands and ensure a reproducible printing process; the influence of printing parameters on extruded strand diameter and cell survival was also investigated. The mechanical strength of the construct was improved by co-printing the fibrin bioink along a supporting PCL carrier scaffold. Compressive mechanical testing showed improved mechanical properties with an average compressive modulus of 131 ± 23 MPa, which falls in the range of cortical bone. HUVEC and hMSC laden fibrin hydrogels were printed in osteon-like patterns and cultured in vitro. A significant increase in gene expression of angiogenic markers was observed for the biomimetic scaffolds. Finally, biphasic scaffolds were implanted subcutaneously in rats. Histological analysis of explanted scaffolds showed a significant increase in the number of blood vessels per area in the 3D printed osteon-like scaffolds. The utilization of these scaffolds in constructing biomimetic osteons for bone regeneration demonstrated a promising capacity to improve neovascularization of the construct. These results indicates that proper cell orientation and scaffold design could play a critical role in neovascularization.
Collapse
Affiliation(s)
- Charlotte Piard
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| | - Hannah Baker
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| | - Timur Kamalitdinov
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| | - John Fisher
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| |
Collapse
|
36
|
Samberg M, Stone R, Natesan S, Kowalczewski A, Becerra S, Wrice N, Cap A, Christy R. Platelet rich plasma hydrogels promote in vitro and in vivo angiogenic potential of adipose-derived stem cells. Acta Biomater 2019; 87:76-87. [PMID: 30665019 DOI: 10.1016/j.actbio.2019.01.039] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/18/2018] [Accepted: 01/17/2019] [Indexed: 12/23/2022]
Abstract
Despite great advances in skin wound care utilizing grafting techniques, the resulting severe scarring, deformity and ineffective vascularization remains a challenge. Alternatively, tissue engineering of new skin using patient-derived stem cells and scaffolding materials promises to greatly increase the functional and aesthetic outcome of skin wound healing. This work focused on the optimization of a polyethylene glycol modified (PEGylated) platelet-rich plasma (PRP) hydrogel for the protracted release of cytokines, growth factors, and signaling molecules and also the delivery of a provisional physical framework for stem cell angiogenesis. Freshly collected whole blood was utilized to synthesize PEGylated PRP hydrogels containing platelet concentrations ranging from 0 to 200,000 platelets/µl. Hydrogels were characterized using thromboelastography and impedance aggregometry for platelet function and were visualized using scanning electron microscopy. To assess the effects of PEGylated PRP hydrogels on cells, PRP solutions were seeded with human adipose-derived stem cells (ASCs) prior to gelation. Following 14 days of incubation in vitro, increased platelet concentrations resulted in higher ASC proliferation and vascular gene and protein expression (assessed via RT-PCR, ELISA, and immunochemistry). Using a rat skin excision model, wounds treated with PRP + ASC hydrogels increased the number of vessels in the wound by day 8 (80.2 vs. 62.6 vessels/mm2) compared to controls. In conclusion, the proposed PEGylated PRP hydrogel promoted both in vitro and transient in vivo angiogenesis of ASCs for improved wound healing. STATEMENT OF SIGNIFICANCE: Our findings support an innovative means of cellular therapy intervention to improve surgical wound healing in a normal wound model. ASCs seeded within PEGylated PRP could be an efficacious and completely autologous therapy for treating patients who have poorly healing wounds caused by vascular insufficiency, previous irradiation, or full-thickness burns. Because wound healing is a dynamic and complex process, the application of more than one growth factor with ASCs demonstrates an advantageous way of improving healing.
Collapse
Affiliation(s)
- Meghan Samberg
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA
| | - Randolph Stone
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA
| | - Shanmugasundaram Natesan
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA
| | - Andrew Kowalczewski
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA
| | - Sandra Becerra
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA
| | - Nicole Wrice
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA
| | - Andrew Cap
- U.S. Army Institute of Surgical Research, Coagulation and Blood Research, JBSA Fort Sam Houston, TX, USA
| | - Robert Christy
- U.S. Army Institute of Surgical Research, Combat Trauma and Burn Injury Research, JBSA Fort Sam Houston, TX, USA.
| |
Collapse
|
37
|
Santos SC, Custódio CA, Mano JF. Photopolymerizable Platelet Lysate Hydrogels for Customizable 3D Cell Culture Platforms. Adv Healthc Mater 2018; 7:e1800849. [PMID: 30387328 DOI: 10.1002/adhm.201800849] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/30/2018] [Indexed: 12/31/2022]
Abstract
3D cell culture platforms have emerged as a setting that resembles in vivo environments replacing the traditional 2D platforms. Over the recent years, an extensive effort has been made on the development of more physiologically relevant 3D cell culture platforms. Extracellular matrix-based materials have been reported as a bioactive and biocompatible support for cell culture. For example, human plasma derivatives have been extensively used in cell culture. Despite all the promising results, in most cases these types of materials have poor mechanical properties and poor stability in vitro. Here plasma-based hydrogels with increased stability are proposed. Platelet lysates are modified by addition of methacryloyl groups (PLMA) that polymerize in controlled geometries upon UV light exposure. The hydrogels could also generate porous scaffolds after lyophilization. The results show that PLMA materials have increased mechanical properties that can be easily adjusted by changing PLMA concentration or modification degree. Cells readily adhere, proliferate, and migrate, exhibiting high viability when encapsulated in PLMA hydrogels. The innovation potential of PLMA materials is based on the fact that it is a complete xeno-free solution for human cell culture, thus an effective alternative to the current gold standards for 3D cell culture based on animal products.
Collapse
Affiliation(s)
- Sara C. Santos
- Department of ChemistryCICECOUniversity of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Catarina A. Custódio
- Department of ChemistryCICECOUniversity of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - João F. Mano
- Department of ChemistryCICECOUniversity of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| |
Collapse
|
38
|
Lin QH, Qu W, Xu J, Feng F, He MF. 1-Methoxycarbony-β-carboline from Picrasma quassioides exerts anti-angiogenic properties in HUVECs in vitro and zebrafish embryos in vivo. Chin J Nat Med 2018; 16:599-609. [PMID: 30197125 DOI: 10.1016/s1875-5364(18)30097-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Indexed: 12/13/2022]
Abstract
Angiogenesis is a crucial process in the development of inflammatory diseases, including cancer, psoriasis and rheumatoid arthritis. Recently, several alkaloids from Picrasma quassioides had been screened for angiogenic activity in the zebrafish model, and the results indicated that 1-methoxycarbony-β-carboline (MCC) could effectively inhibit blood vessel formation. In this study, we further confirmed that MCC can inhibit, in a concentration-dependent manner, the viability, migration, invasion, and tube formation of human umbilical vein endothelial cells (HUVECs) in vitro, as well as the regenerative vascular outgrowth of zebrafish caudal fin in vivo. In the zebrafish xenograft assay, MCC inhibited the growth of tumor masses and the metastatic transplanted DU145 tumor cells. The proteome profile array of the MCC-treated HUVECs showed that MCC could down-regulate several angiogenesis-related self-secreted proteins, including ANG, EGF, bFGF, GRO, IGF-1, PLG and MMP-1. In addition, the expression of two key membrane receptor proteins in angiogenesis, TIE-2 and uPAR, were also down-regulated after MCC treatment. Taken together, these results shed light on the potential therapeutic application of MCC as a potent natural angiogenesis inhibitor via multiple molecular targets.
Collapse
Affiliation(s)
- Qing-Hua Lin
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jian Xu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 211198, China.
| | - Ming-Fang He
- Institute of Translational Medicine, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211800, China.
| |
Collapse
|
39
|
Ke D, Murphy SV. Current Challenges of Bioprinted Tissues Toward Clinical Translation. TISSUE ENGINEERING PART B-REVIEWS 2018; 25:1-13. [PMID: 30129878 DOI: 10.1089/ten.teb.2018.0132] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IMPACT STATEMENT This review has a broad overview of the current challenges of bioprinted tissues towards clinical translations and future directions to overcome those challenges. The development of this field has a huge impact on the situation of an insufficient number of organ donors for life-saving organ transplantations.
Collapse
Affiliation(s)
- Dongxu Ke
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
40
|
Schöneberg J, De Lorenzi F, Theek B, Blaeser A, Rommel D, Kuehne AJC, Kießling F, Fischer H. Engineering biofunctional in vitro vessel models using a multilayer bioprinting technique. Sci Rep 2018; 8:10430. [PMID: 29992981 PMCID: PMC6041340 DOI: 10.1038/s41598-018-28715-0] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/27/2018] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of bioprinting have led to the development of perfusable complex structures. However, most of the existing printed vascular channels lack the composition or key structural and physiological features of natural blood vessels or they make use of more easily printable but less biocompatible hydrogels. Here, we use a drop-on-demand bioprinting technique to generate in vitro blood vessel models, consisting of a continuous endothelium imitating the tunica intima, an elastic smooth muscle cell layer mimicking the tunica media, and a surrounding fibrous and collagenous matrix of fibroblasts mimicking the tunica adventitia. These vessel models with a wall thickness of up to 425 µm and a diameter of about 1 mm were dynamically cultivated in fluidic bioreactors for up to three weeks under physiological flow conditions. High cell viability (>83%) after printing and the expression of VE-Cadherin, smooth muscle actin, and collagen IV were observed throughout the cultivation period. It can be concluded that the proposed novel technique is suitable to achieve perfusable vessel models with a biofunctional multilayer wall composition. Such structures hold potential for the creation of more physiologically relevant in vitro disease models suitable especially as platforms for the pre-screening of drugs.
Collapse
Affiliation(s)
- Jan Schöneberg
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Federica De Lorenzi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Benjamin Theek
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Blaeser
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Dirk Rommel
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen, Germany
| | - Alexander J C Kuehne
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen, Germany
| | - Fabian Kießling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
41
|
Huang Q, Zou Y, Arno MC, Chen S, Wang T, Gao J, Dove AP, Du J. Hydrogel scaffolds for differentiation of adipose-derived stem cells. Chem Soc Rev 2018; 46:6255-6275. [PMID: 28816316 DOI: 10.1039/c6cs00052e] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural extracellular matrices (ECMs) have been widely used as a support for the adhesion, migration, differentiation, and proliferation of adipose-derived stem cells (ADSCs). However, poor mechanical behavior and unpredictable biodegradation properties of natural ECMs considerably limit their potential for bioapplications and raise the need for different, synthetic scaffolds. Hydrogels are regarded as the most promising alternative materials as a consequence of their excellent swelling properties and their resemblance to soft tissues. A variety of strategies have been applied to create synthetic biomimetic hydrogels, and their biophysical and biochemical properties have been modulated to be suitable for cell differentiation. In this review, we first give an overview of common methods for hydrogel preparation with a focus on those strategies that provide potential advantages for ADSC encapsulation, before summarizing the physical properties of hydrogel scaffolds that can act as biological cues. Finally, the challenges in the preparation and application of hydrogels with ADSCs are explored and the perspectives are proposed for the next generation of scaffolds.
Collapse
Affiliation(s)
- Qiutong Huang
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai, 201804, China.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 347] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
43
|
Injectable chitosan-fibrin/nanocurcumin composite hydrogel for the enhancement of angiogenesis. RESEARCH ON CHEMICAL INTERMEDIATES 2018. [DOI: 10.1007/s11164-018-3340-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
44
|
Manavella DD, Cacciottola L, Desmet CM, Jordan BF, Donnez J, Amorim CA, Dolmans MM. Adipose tissue-derived stem cells in a fibrin implant enhance neovascularization in a peritoneal grafting site: a potential way to improve ovarian tissue transplantation. Hum Reprod 2018; 33:270-279. [DOI: 10.1093/humrep/dex374] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/11/2017] [Indexed: 12/12/2022] Open
|
45
|
Aurora A, Wrice N, Walters TJ, Christy RJ, Natesan S. A PEGylated platelet free plasma hydrogel based composite scaffold enables stable vascularization and targeted cell delivery for volumetric muscle loss. Acta Biomater 2018; 65:150-162. [PMID: 29128541 DOI: 10.1016/j.actbio.2017.11.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 10/26/2017] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
Abstract
Extracellular matrix (ECM) scaffolds are being used for the clinical repair of soft tissue injuries. Although improved functional outcomes have been reported, ECM scaffolds show limited tissue specific remodeling response with concomitant deposition of fibrotic tissue. One plausible explanation is the regression of blood vessels which may be limiting the diffusion of oxygen and nutrients across the scaffold. Herein we develop a composite scaffold as a vasculo-inductive platform by integrating PEGylated platelet free plasma (PFP) hydrogel with a muscle derived ECM scaffold (m-ECM). In vitro, adipose derived stem cells (ASCs) seeded onto the composite scaffold differentiated into two distinct morphologies, a tubular network in the hydrogel, and elongated structures along the m-ECM scaffold. The composite scaffold showed a high expression of ITGA5, ITGB1, and FN and a synergistic up-regulation of ang1 and tie-2 transcripts. The in vitro ability of the composite scaffold to provide extracellular milieu for cell adhesion and molecular cues to support vessel formation was investigated in a rodent volumetric muscle loss (VML) model. The composite scaffold delivered with ASCs supported robust and stable vascularization. Additionally, the composite scaffold supported increased localization of ASCs in the defect demonstrating its ability for localized cell delivery. Interestingly, ASCs were observed homing in the injured muscle and around the perivascular space possibly to stabilize the host vasculature. In conclusion, the composite scaffold delivered with ASCs presents a promising approach for scaffold vascularization. The versatile nature of the composite scaffold also makes it easily adaptable for the repair of soft tissue injuries. STATEMENT OF SIGNIFICANCE Decellularized extracellular matrix (ECM) scaffolds when used for soft tissue repair is often accompanied by deposition of fibrotic tissue possibly due to limited scaffold vascularization, which limits the diffusion of oxygen and nutrients across the scaffold. Although a variety of scaffold vascularization strategies has been investigated, their limitations preclude rapid clinical translation. In this study we have developed a composite scaffold by integrating bi-functional polyethylene glycol modified platelet free plasma (PEGylated PFP) with adipose derived stem cells (ASCs) along with a muscle derived ECM scaffold (m-ECM). The composite scaffold provides a vasculo-inductive and an effective cell delivery platform for volumetric muscle loss.
Collapse
|
46
|
Xue A, Niu G, Chen Y, Li K, Xiao Z, Luan Y, Sun C, Xie X, Zhang D, Du X, Kong F, Guo Y, Zhang H, Cheng G, Xin Q, Guan Y, Zhao S. Recellularization of well-preserved decellularized kidney scaffold using adipose tissue-derived stem cells. J Biomed Mater Res A 2017; 106:805-814. [PMID: 29067774 DOI: 10.1002/jbm.a.36279] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/29/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Aibing Xue
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Guangzhu Niu
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Yuan Chen
- Department of Central Research Lab; The Second Hospital, Shandong University; Jinan Shandong China
| | - Kailin Li
- Department of Central Research Lab; The Second Hospital, Shandong University; Jinan Shandong China
| | - Zhiying Xiao
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Yun Luan
- Department of Central Research Lab; The Second Hospital, Shandong University; Jinan Shandong China
| | - Chao Sun
- Department of Central Research Lab; The Second Hospital, Shandong University; Jinan Shandong China
| | - Xiaoshuai Xie
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Denglu Zhang
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Xiaohang Du
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Feng Kong
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Yanxia Guo
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Haiyang Zhang
- Minimally Invasive Urology Center, Shandong Provincial Hospital affiliated to Shandong University; Jinan Shandong China
| | - Guanghui Cheng
- Department of Central Research Lab; The Second Hospital, Shandong University; Jinan Shandong China
| | - Qian Xin
- Department of Central Research Lab; The Second Hospital, Shandong University; Jinan Shandong China
| | - Yong Guan
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
| | - Shengtian Zhao
- Department of Urology; The Second Hospital, Shandong University; Jinan Shandong China
- Department of Urology; The Affiliated Hospital of Shandong University of Traditional Chinese Medicine; Jinan Shandong China
| |
Collapse
|
47
|
Kuss MA, Wu S, Wang Y, Untrauer JB, Li W, Lim JY, Duan B. Prevascularization of 3D printed bone scaffolds by bioactive hydrogels and cell co-culture. J Biomed Mater Res B Appl Biomater 2017; 106:1788-1798. [PMID: 28901689 DOI: 10.1002/jbm.b.33994] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/25/2017] [Accepted: 08/28/2017] [Indexed: 01/11/2023]
Abstract
Vascularization is a fundamental prerequisite for large bone construct development and remains one of the main challenges of bone tissue engineering. Our current study presents the combination of 3D printing technique with a hydrogel-based prevascularization strategy to generate prevascularized bone constructs. Human adipose derived mesenchymal stem cells (ADMSC) and human umbilical vein endothelial cells (HUVEC) were encapsulated within our bioactive hydrogels, and the effects of culture conditions on in vitro vascularization were determined. We further generated composite constructs by forming 3D printed polycaprolactone/hydroxyapatite scaffolds coated with cell-laden hydrogels and determined how the co-culture affected vascularization and osteogenesis. It was demonstrated that 3D co-cultured ADMSC-HUVEC generated capillary-like networks within the porous 3D printed scaffold. The co-culture systems promoted in vitro vascularization, but had no significant effects on osteogenesis. The prevascularized constructs were subcutaneously implanted into nude mice to evaluate the in vivo vascularization capacity and the functionality of engineered vessels. The hydrogel systems facilitated microvessel and lumen formation and promoted anastomosis of vascular networks of human origin with host murine vasculature. These findings demonstrate the potential of prevascularized 3D printed scaffolds with anatomical shape for the healing of larger bone defects. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1788-1798, 2018.
Collapse
Affiliation(s)
- Mitchell A Kuss
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Shaohua Wu
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ying Wang
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jason B Untrauer
- Division of Oral and Maxillofacial Surgery, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Wenlong Li
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Jung Yul Lim
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
48
|
Abstract
OBJECTIVE A vibratory vocal fold replacement would introduce a new treatment paradigm for structural vocal fold diseases such as scarring and lamina propria loss. This work implants a tissue-engineered replacement for vocal fold lamina propria and epithelium in rabbits and compares histology and function to injured controls and orthotopic transplants. Hypotheses were that the cell-based implant would engraft and control the wound response, reducing fibrosis and restoring vibration. STUDY DESIGN Translational research. METHODS Rabbit adipose-derived mesenchymal stem cells (ASC) were embedded within a three-dimensional fibrin gel, forming the cell-based outer vocal fold replacement (COVR). Sixteen rabbits underwent unilateral resection of vocal fold epithelium and lamina propria, as well as reconstruction with one of three treatments: fibrin glue alone with healing by secondary intention, replantation of autologous resected vocal fold cover, or COVR implantation. After 4 weeks, larynges were examined histologically and with phonation. RESULTS Fifteen rabbits survived. All tissues incorporated well after implantation. After 1 month, both graft types improved histology and vibration relative to injured controls. Extracellular matrix (ECM) of the replanted mucosa was disrupted, and ECM of the COVR implants remained immature. Immune reaction was evident when male cells were implanted into female rabbits. Best histologic and short-term vibratory outcomes were achieved with COVR implants containing male cells implanted into male rabbits. CONCLUSION Vocal fold cover replacement with a stem cell-based tissue-engineered construct is feasible and beneficial in acute rabbit implantation. Wound-modifying behavior of the COVR implant is judged to be an important factor in preventing fibrosis. LEVEL OF EVIDENCE NA. Laryngoscope, 128:153-159, 2018.
Collapse
Affiliation(s)
- Jennifer L Long
- Research Service, Greater Los Angeles VAHS, Los Angeles, California, U.S.A.,Department of Head and Neck Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, U.S.A
| |
Collapse
|
49
|
|
50
|
Gulati K, Meher MK, Poluri KM. Glycosaminoglycan-based resorbable polymer composites in tissue refurbishment. Regen Med 2017. [DOI: 10.2217/rme-2017-0012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Regeneration of tissue structure with the aid of bioactive polymer matrices/composites and scaffolds for respective applications is one of the emerging areas of biomedical engineering. Recent advances in conjugated glycosaminoglycan (GAG) hybrids using natural and synthetic polymers have opened new avenues for producing a wide variety of resorbable polymer matrices. These hybrid scaffolds are low-immunogenic, highly biocompatible and biodegradable with incredible mechanical and tensile properties. GAG-based resorbable polymeric matrices are being exploited in migration of stem cells, cartilage and bone replacement/regeneration and production of scaffolds for various tissue engineering applications. In the current review, we will discuss the role of GAG-based resorbable polymer matrices in the field of regenerative medicine.
Collapse
Affiliation(s)
- Khushboo Gulati
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Mukesh Kumar Meher
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|