1
|
Mansouri Moghaddam M, Jooybar E, Imani R. Injectable microgel and micro-granular hydrogels for bone tissue engineering. Biofabrication 2025; 17:032001. [PMID: 40228520 DOI: 10.1088/1758-5090/adcc58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/14/2025] [Indexed: 04/16/2025]
Abstract
Injectable microgels, made from both natural and synthetic materials, are promising platforms for the encapsulation of cells or bioactive agents, such as drugs and growth factors, for delivery to injury sites. They can also serve as effective micro-scaffolds in bone tissue engineering (BTE), offering a supportive environment for cell proliferation or differentiation into osteoblasts. Microgels can be injected in the injury sites individually or in the form of aggregated/jammed ones named micro-granular hydrogels. This review focuses on common materials and fabrication techniques for preparing injectable microgels, as well as their characteristics and applications in BTE. These applications include their use as cell carriers, delivery systems for bioactive molecules, micro-granular hydrogels, bio-inks for bioprinting, three-dimensional microarrays, and the formation of microtissues. Furthermore, we discuss the current and potential future applications of microgels in bone tissue regeneration.
Collapse
Affiliation(s)
- Melika Mansouri Moghaddam
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
2
|
Wei C, Guo Y, Ci Z, Li M, Zhang Y, Zhou Y. Advances of Schwann cells in peripheral nerve regeneration: From mechanism to cell therapy. Biomed Pharmacother 2024; 175:116645. [PMID: 38729050 DOI: 10.1016/j.biopha.2024.116645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Peripheral nerve injuries (PNIs) frequently occur due to various factors, including mechanical trauma such as accidents or tool-related incidents, as well as complications arising from diseases like tumor resection. These injuries frequently result in persistent numbness, impaired motor and sensory functions, neuropathic pain, or even paralysis, which can impose a significant financial burden on patients due to outcomes that often fall short of expectations. The most frequently employed clinical treatment for PNIs involves either direct sutures of the severed ends or bridging the proximal and distal stumps using autologous nerve grafts. However, autologous nerve transplantation may result in sensory and motor functional loss at the donor site, as well as neuroma formation and scarring. Transplantation of Schwann cells/Schwann cell-like cells has emerged as a promising cellular therapy to reconstruct the microenvironment and facilitate peripheral nerve regeneration. In this review, we summarize the role of Schwann cells and recent advances in Schwann cell therapy in peripheral nerve regeneration. We summarize current techniques used in cell therapy, including cell injection, 3D-printed scaffolds for cell delivery, cell encapsulation techniques, as well as the cell types employed in experiments, experimental models, and research findings. At the end of the paper, we summarize the challenges and advantages of various cells (including ESCs, iPSCs, and BMSCs) in clinical cell therapy. Our goal is to provide the theoretical and experimental basis for future treatments targeting peripheral nerves, highlighting the potential of cell therapy and tissue engineering as invaluable resources for promoting nerve regeneration.
Collapse
Affiliation(s)
- Chuqiao Wei
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuanxin Guo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhen Ci
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Mucong Li
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China.
| |
Collapse
|
3
|
Chen X, Huang S, Niu Y, Luo M, Liu H, Jiao Y, Huang J. Transplantation of Gelatin Microspheres Loaded with Wharton's Jelly Derived Mesenchymal Stem Cells Facilitates Cartilage Repair in Mice. Tissue Eng Regen Med 2024; 21:171-183. [PMID: 37688747 PMCID: PMC10764672 DOI: 10.1007/s13770-023-00574-5] [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: 04/18/2023] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a prevalent chronic joint disease caused by various factors. Mesenchymal stem cells (MSCs) therapy is an increasingly promising therapeutic option for osteoarthritis. However, the chronic inflammation of knee joint can severely impede the therapeutic effects of transplanted cells. Gelatin microspheres (GMs) are degradable biomaterial that have various porosities for cell adhesion and cell-cell interaction. Excellent elasticity and deformability of GMs make it an excellent injectable vehicle for cell delivery. METHODS We created Wharton's jelly derived mesenchymal stem cells (WJMSCs)-GMs complexes and assessed the effects of GMs on cell activity, proliferation and chondrogenesis. Then, WJMSCs loaded in GMs were transplanted in the joint of osteoarthritis mice. After four weeks, joint tissue was collected for histological analysis. Overexpressing-luciferase WJMSCs were performed to explore cell retention in mice. RESULTS In vitro experiments demonstrated that WJMSCs loaded with GMs maintained cell viability and proliferative potential. Moreover, GMs enhanced the chondrogenesis differentiation of WJMSCs while alleviated cell hypertrophy. In KOA mice model, transplantation of WJMSCs-GMs complexes promoted cartilage regeneration and cartilage matrix formation, contributing to the treatment of KOA. Compared with other groups, in WJMSCs+GMs group, there were fewer cartilage defects and with a more integrated tibia structure. Tracking results of stable-overexpressing luciferase WJMSCs demonstrated that GMs significantly extended the retention time of WJMSCs in knee joint cavity. CONCLUSION Our results indicated that GMs facilitate WJMSCs mediated knee osteoarthritis healing in mice by promoting cartilage regeneration and prolonging cell retention. It might potentially provide an optimal strategy for the biomaterial-stem cell based therapy for knee osteoarthritis.
Collapse
Affiliation(s)
- Xiaolin Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Sunxing Huang
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affliated Hospital and School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yongxia Niu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Mingxun Luo
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Haiying Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yiren Jiao
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affliated Hospital and School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
4
|
Kim B, Lee B, Mandakhbayar N, Kim Y, Song Y, Doh J, Lee JH, Jeong B, Song KH. Effect of lyophilized gelatin-norbornene cryogel size on calvarial bone regeneration. Mater Today Bio 2023; 23:100868. [PMID: 38075253 PMCID: PMC10701364 DOI: 10.1016/j.mtbio.2023.100868] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/26/2023] [Accepted: 11/14/2023] [Indexed: 04/09/2025] Open
Abstract
Molding processes with molds containing topographical structures have been used for fabrication of hydrogel and cryogel particles. However, they can involve difficulties in separation of fabricated particles with complex shape from the molds or repeated fabrication of the particles although the overall processes do not require much skill and equipment. In this study, molds with etched superhydrophobic patterns have been developed by etching polytetrafluoroethylene (PTFE) blocks in user-defined designs with a femtosecond (FS) laser-based etching system. Lyophilized cryogel particles with various designs and sizes were fabricated by molding precursors with these PTFE molds. Additionally, the clean and easy separation of particles from the molds allowed repeated fabrication of the particles. For an application, relatively 'big' gelatin-norbornene (GelNB) cryogel particles prepared via molding with polydimethylsiloxane (PDMS) molds, swelling in phosphate buffered saline (PBS) and slicing height in half and 'small' GelNB cryogel particles fabricated with the PTFE molds were fabricated. Then, they were used to study scaffold size effect on calvarial bone regeneration. The molds generated with the FS laser-based etching system can be useful for various applications that require the mass production of cryogel particles in various geometries.
Collapse
Affiliation(s)
- Bomi Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Byunghak Lee
- B2LAB Co., Ltd., Seoul, 05836, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210, Mongolia
| | | | - Yeonju Song
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Junsang Doh
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research: SOFT Foundry Institute, Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Bosu Jeong
- B2LAB Co., Ltd., Seoul, 05836, Republic of Korea
| | - Kwang Hoon Song
- Department of Nano-Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, 22012, Republic of Korea
| |
Collapse
|
5
|
Hamilton M, Wang J, Dhar P, Stehno-Bittel L. Controlled-Release Hydrogel Microspheres to Deliver Multipotent Stem Cells for Treatment of Knee Osteoarthritis. Bioengineering (Basel) 2023; 10:1315. [PMID: 38002439 PMCID: PMC10669156 DOI: 10.3390/bioengineering10111315] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Osteoarthritis (OA) is the most common form of joint disease affecting articular cartilage and peri-articular tissues. Traditional treatments are insufficient, as they are aimed at mitigating symptoms. Multipotent Stromal Cell (MSC) therapy has been proposed as a treatment capable of both preventing cartilage destruction and treating symptoms. While many studies have investigated MSCs for treating OA, therapeutic success is often inconsistent due to low MSC viability and retention in the joint. To address this, biomaterial-assisted delivery is of interest, particularly hydrogel microspheres, which can be easily injected into the joint. Microspheres composed of hyaluronic acid (HA) were created as MSC delivery vehicles. Microrheology measurements indicated that the microspheres had structural integrity alongside sufficient permeability. Additionally, encapsulated MSC viability was found to be above 70% over one week in culture. Gene expression analysis of MSC-identifying markers showed no change in CD29 levels, increased expression of CD44, and decreased expression of CD90 after one week of encapsulation. Analysis of chondrogenic markers showed increased expressions of aggrecan (ACAN) and SRY-box transcription factor 9 (SOX9), and decreased expression of osteogenic markers, runt-related transcription factor 2 (RUNX2), and alkaline phosphatase (ALPL). In vivo analysis revealed that HA microspheres remained in the joint for up to 6 weeks. Rats that had undergone destabilization of the medial meniscus and had overt OA were treated with empty HA microspheres, MSC-laden microspheres, MSCs alone, or a control vehicle. Pain measurements taken before and after the treatment illustrated temporarily decreased pain in groups treated with encapsulated cells. Finally, the histopathological scoring of each group illustrated significantly less OA damage in those treated with encapsulated cells compared to controls. Overall, these studies demonstrate the potential of using HA-based hydrogel microspheres to enhance the therapeutic efficacy of MSCs in treating OA.
Collapse
Affiliation(s)
- Megan Hamilton
- Bioengineering Program, School of Engineering, University of Kansas, Lawrence, KS 66045, USA;
- Likarda, Kansas City, MO 64137, USA;
| | - Jinxi Wang
- Department of Orthopedic Surgery and Sport Medicine, School of Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Prajnaparamita Dhar
- Bioengineering Program, School of Engineering, University of Kansas, Lawrence, KS 66045, USA;
| | - Lisa Stehno-Bittel
- Likarda, Kansas City, MO 64137, USA;
- Department of Orthopedic Surgery and Sport Medicine, School of Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| |
Collapse
|
6
|
Lee SS, Kleger N, Kuhn GA, Greutert H, Du X, Smit T, Studart AR, Ferguson SJ. A 3D-Printed Assemblable Bespoke Scaffold as a Versatile Microcryogel Carrier for Site-Specific Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302008. [PMID: 37632210 DOI: 10.1002/adma.202302008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/22/2023] [Indexed: 08/27/2023]
Abstract
Advances in additive manufacturing have led to diverse patient-specific implant designs utilizing computed tomography, but this requires intensive work and financial implications. Here, Digital Light Processing is used to fabricate a hive-structured assemblable bespoke scaffold (HIVE). HIVE can be manually assembled in any shape/size with ease, so a surgeon can create a scaffold that will best fit a defect before implantation. Simultaneously, it can have site-specific treatments by working as a carrier filled with microcryogels (MC) incorporating different biological factors in different pockets of HIVE. After characterization, possible site-specific applications are investigated by utilizing HIVE as a versatile carrier with incorporated treatments such as growth factors (GF), bioceramic, or cells. HIVE as a GF-carrier shows a controlled release of bone morphogenetic protein/vascular endothelial growth factor (BMP/VEGF) and induced osteogenesis/angiogenesis from human mesenchymal stem cells (hMSC)/human umbilical vein endothelial cells (HUVECs). Furthermore, as a bioceramic-carrier, HIVE demonstrates enhanced mineralization and osteogenesis, and as a HUVEC carrier, it upregulates both osteogenic and angiogenic gene expression of hMSCs. HIVE with different combinations of MCs yields a distinct local effect and successful cell migration is confirmed within assembled HIVEs. Finally, an in vivo rat subcutaneous implantation demonstrates site-specific osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- Seunghun S Lee
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Nicole Kleger
- Complex Materials, Department of Materials, ETH Zurich, Zurich, 8093, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Helen Greutert
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Xiaoyu Du
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - Thijs Smit
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| | - André R Studart
- Complex Materials, Department of Materials, ETH Zurich, Zurich, 8093, Switzerland
| | - Stephen J Ferguson
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, 8092, Switzerland
| |
Collapse
|
7
|
Yang Z, Wang B, Liu W, Li X, Liang K, Fan Z, Li JJ, Niu Y, He Z, Li H, Wang D, Lin J, Du Y, Lin J, Xing D. In situ self-assembled organoid for osteochondral tissue regeneration with dual functional units. Bioact Mater 2023; 27:200-215. [PMID: 37096194 PMCID: PMC10121637 DOI: 10.1016/j.bioactmat.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/01/2023] [Accepted: 04/02/2023] [Indexed: 04/26/2023] Open
Abstract
The regeneration of hierarchical osteochondral units is challenging due to difficulties in inducing spatial, directional and controllable differentiation of mesenchymal stem cells (MSCs) into cartilage and bone compartments. Emerging organoid technology offers new opportunities for osteochondral regeneration. In this study, we developed gelatin-based microcryogels customized using hyaluronic acid (HA) and hydroxyapatite (HYP), respectively for inducing cartilage and bone regeneration (denoted as CH-Microcryogels and OS-Microcryogels) through in vivo self-assembly into osteochondral organoids. The customized microcryogels showed good cytocompatibility and induced chondrogenic and osteogenic differentiation of MSCs, while also demonstrating the ability to self-assemble into osteochondral organoids with no delamination in the biphasic cartilage-bone structure. Analysis by mRNA-seq showed that CH-Microcryogels promoted chondrogenic differentiation and inhibited inflammation, while OS-Microcryogels facilitated osteogenic differentiation and suppressed the immune response, by regulating specific signaling pathways. Finally, the in vivo engraftment of pre-differentiated customized microcryogels into canine osteochondral defects resulted in the spontaneous assembly of an osteochondral unit, inducing simultaneous regeneration of both articular cartilage and subchondral bone. In conclusion, this novel approach for generating self-assembling osteochondral organoids utilizing tailor-made microcryogels presents a highly promising avenue for advancing the field of tissue engineering.
Collapse
Affiliation(s)
- Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Bin Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Xiaoke Li
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zejun Fan
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, Australia
| | - Yudi Niu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zihao He
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Hui Li
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Jianhao Lin
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Dan Xing
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| |
Collapse
|
8
|
Wang Y, Yuan Z, Pang Y, Zhang D, Li G, Zhang X, Yu Y, Yang X, Cai Q. Injectable, High Specific Surface Area Cryogel Microscaffolds Integrated with Osteoinductive Bioceramic Fibers for Enhanced Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20661-20676. [PMID: 37083252 DOI: 10.1021/acsami.3c00192] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Organic-inorganic composites with high specific surface area and osteoinductivity provide a suitable microenvironment for cell ingrowth and effective ossification, which could greatly promote bone regeneration. Here, we report gelatin methacryloyl (GelMA) cryogel microspheres that are reinforced with hydroxyapatite (HA) nanowires and calcium silicate (CS) nanofibers to achieve the goal. The prepared composite cryogel microspheres with open porous structure and rough surface greatly facilitate cell anchoring, simultaneously exhibiting excellent injectability. Compared to the only HA- or CS-containing counterparts, the GelMA cryogel microspheres composited with HA:CS (termed as GMHC) achieve sustained release of bioactive Ca, P, and Si elements, which are conducive to osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs). These composite microspheres can prevent from forming peralkalic conditions, which is beneficial for cell growth. After injection of cryogel microspheres into rat calvarial defects, neo-bone tissue grows into their pores, showing tight integration. The embedded bioceramic components significantly promote bone regeneration, with the GMHC achieving the best regenerative outcomes. Promisingly, porous organic-inorganic composite cryogel microspheres, with high specific surface area, biodegradability, and osteoinductivity, can act as injectable microscaffolds to repair bone defects with enhanced efficiency, which may widen the scaffold strategy for bone tissue engineering.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Yanyun Pang
- School of Stomatology, Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Daixing Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Guangyu Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xu Zhang
- School of Stomatology, Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoping Yang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
9
|
Özbek MA, Bereli N, Özgür E, Denizli A. Human serum albumin depletion based on dye ligand affinity chromatography via magnetic microcryogels. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:449-462. [PMID: 36123323 DOI: 10.1080/09205063.2022.2127142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
One of the primary purposes of proteomic studies is to analyze the proteins in the blood to be considered as biomarkers. Albumin, which constitutes the majority of total serum proteins, complicates the discovery of low-density proteins that are important for the diagnosis of diseases. Based on this, an alternative approach for albumin depletion was developed in this study by covalently attached Cibacron Blue 3GA (CB) to magnetic microcryogels. After detailed characterization of CB attached magnetic microcryogels synthesized via a microstencil array chip, albumin adsorption studies were performed to examine the optimum depletion conditions. In the presented study, the maximum albumin adsorption capacity (Qmax) was calculated as 149.25 mg/mL in pH 5.0 acetate buffer solution, which is the optimum pH value for albumin. Experimental studies have demonstrated that CB-attached magnetic microcryogels can be reused without loss of performance for albumin depletion after 10 adsorption-desorption cycles.
Collapse
Affiliation(s)
- Merve Asena Özbek
- Faculty of Science, Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Nilay Bereli
- Faculty of Science, Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Erdoğan Özgür
- Faculty of Science, Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Adil Denizli
- Faculty of Science, Department of Chemistry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
10
|
Ding SL, Liu X, Zhao XY, Wang KT, Xiong W, Gao ZL, Sun CY, Jia MX, Li C, Gu Q, Zhang MZ. Microcarriers in application for cartilage tissue engineering: Recent progress and challenges. Bioact Mater 2022; 17:81-108. [PMID: 35386447 PMCID: PMC8958326 DOI: 10.1016/j.bioactmat.2022.01.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Successful regeneration of cartilage tissue at a clinical scale has been a tremendous challenge in the past decades. Microcarriers (MCs), usually used for cell and drug delivery, have been studied broadly across a wide range of medical fields, especially the cartilage tissue engineering (TE). Notably, microcarrier systems provide an attractive method for regulating cell phenotype and microtissue maturations, they also serve as powerful injectable carriers and are combined with new technologies for cartilage regeneration. In this review, we introduced the typical methods to fabricate various types of microcarriers and discussed the appropriate materials for microcarriers. Furthermore, we highlighted recent progress of applications and general design principle for microcarriers. Finally, we summarized the current challenges and promising prospects of microcarrier-based systems for medical applications. Overall, this review provides comprehensive and systematic guidelines for the rational design and applications of microcarriers in cartilage TE.
Collapse
Affiliation(s)
- Sheng-Long Ding
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xin Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xi-Yuan Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ke-Tao Wang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Wei Xiong
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zi-Li Gao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Cheng-Yi Sun
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Min-Xuan Jia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Cheng Li
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ming-Zhu Zhang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
11
|
Li Y, Xu W, Li Q, Li X, Li J, Kang L, Fang Y, Cheng S, Zhao P, Jiang S, Liu W, Yan X, Du Y, Wang L, Huang Y. 3D biomaterial P scaffolds carrying umbilical cord mesenchymal stem cells improve biointegration of keratoprosthesis. Biomed Mater 2022; 17. [PMID: 35790151 DOI: 10.1088/1748-605x/ac7e98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/05/2022] [Indexed: 11/12/2022]
Abstract
Biointegration of a keratoprosthesis (KPro) is critical for the device stability and long-term retention. Biointegration of the KPro device and host tissue takes place between the surrounding corneal graft and the central optic [made by poly (methyl methacrylate) (PMMA)]. Our previous clinical results showed that auricular cartilage reinforcement is able to enhance the KPro biointegration. However, the auricular cartilage is non-renewable and difficult to acquire. In this study, we developed a novel type of biomaterial using a three-dimensional porous polyethylene glycol acrylate scaffold (3D biological P-scaffold) carrier with chondrocytes differentiated from induced human umbilical cord mesenchymal stem cells (hUC-MSCs) and tested in rabbit corneas. The results showed hUC-MSCs bear stem cell properties and coule be induced into chondrocytes, P-scaffold is beneficial to the growth and differentiation of hUC-MSCs both in vivo and in vitro. Besides, after implanting the P-scaffold into the corneal stroma, no serious immune rejection response, such as corneal ulcer or perforation were seen, suggested a good biocompatibility of P-scaffold with the corneal tissue. Moreover, after implanting P-scaffold in together with the differentiated chondrocytes into the rabbit corneal stroma, they significantly increased corneal thickness and strengthened the host cornea, and chondrocytes could stably persist inside the cornea. In summary, the 3D biological P-scaffold carrying differentiated hUC-MSCs could be the preferable material for KPro reinforcement.
Collapse
Affiliation(s)
- Yueyue Li
- Medical School of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Wenqin Xu
- Ophthalmology, General Hospital of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Qian Li
- Medical School of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Xiaoqi Li
- Medical School of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Junyang Li
- Biomedical Engineering, Tsinghua University School of Medicine, Shuangqing Road 30, Beijing, Beijing, 100084, CHINA
| | - Li Kang
- Medical School of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Yifan Fang
- Medical School of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Shuaishuai Cheng
- Biomedical Engineering, Tsinghua University School of Medicine, Shuangqing Road 30, Beijing, Beijing, 100084, CHINA
| | - Peng Zhao
- Biomedical Engineering, Tsinghua University School of Medicine, Shuangqing Road 30, Beijing, Beijing, 100084, CHINA
| | - Shumeng Jiang
- Biomedical Engineering, Tsinghua University School of Medicine, Shuangqing Road 30, Beijing, Beijing, 100084, CHINA
| | - Wei Liu
- Biomedical Engineering, Tsinghua University School of Medicine, Shuangqing Road 30, Beijing, Beijing, 100084, CHINA
| | - Xiaojun Yan
- Biomedical Engineering, Tsinghua University School of Medicine, Shuangqing Road 30, Beijing, Beijing, 100084, CHINA
| | - Yanan Du
- Tsinghua University, China-Beijing-Tsinghua University, Beijing, 100084, CHINA
| | - Liqiang Wang
- Ophthalmology, General Hospital of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| | - Yifei Huang
- Ophthalmology, General Hospital of People's Liberation Army, Fuxing Road 28, Beijing, 100853, CHINA
| |
Collapse
|
12
|
Xu F, Dawson C, Lamb M, Mueller E, Stefanek E, Akbari M, Hoare T. Hydrogels for Tissue Engineering: Addressing Key Design Needs Toward Clinical Translation. Front Bioeng Biotechnol 2022; 10:849831. [PMID: 35600900 PMCID: PMC9119391 DOI: 10.3389/fbioe.2022.849831] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Graphical Abstract
Collapse
Affiliation(s)
- Fei Xu
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Chloe Dawson
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Makenzie Lamb
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Eva Mueller
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Evan Stefanek
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC, Canada
| | - Mohsen Akbari
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC, Canada
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
- *Correspondence: Mohsen Akbari, ; Todd Hoare,
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
- *Correspondence: Mohsen Akbari, ; Todd Hoare,
| |
Collapse
|
13
|
Çetin K, Denizli A. Polyethylenimine-functionalized microcryogels for controlled release of diclofenac sodium. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2021.105125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
14
|
|
15
|
Wang B, Liu W, Li JJ, Chai S, Xing D, Yu H, Zhang Y, Yan W, Xu Z, Zhao B, Du Y, Jiang Q. A low dose cell therapy system for treating osteoarthritis: In vivo study and in vitro mechanistic investigations. Bioact Mater 2022; 7:478-490. [PMID: 34466747 PMCID: PMC8379370 DOI: 10.1016/j.bioactmat.2021.05.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be effective in alleviating the progression of osteoarthritis (OA). However, low MSC retention and survival at the injection site frequently require high doses of cells and/or repeated injections, which are not economically viable and create additional risks of complications. In this study, we produced MSC-laden microcarriers in spinner flask culture as cell delivery vehicles. These microcarriers containing a low initial dose of MSCs administered through a single injection in a rat anterior cruciate ligament (ACL) transection model of OA achieved similar reparative effects as repeated high doses of MSCs, as evaluated through imaging and histological analyses. Mechanistic investigations were conducted using a co-culture model involving human primary chondrocytes grown in monolayer, together with MSCs grown either within 3D constructs or as a monolayer. Co-culture supernatants subjected to secretome analysis showed significant decrease of inflammatory factors in the 3D group. RNA-seq of co-cultured MSCs and chondrocytes using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed processes relating to early chondrogenesis and increased extracellular matrix interactions in MSCs of the 3D group, as well as phenotypic maintenance in the co-cultured chondrocytes. The cell delivery platform we investigated may be effective in reducing the cell dose and injection frequency required for therapeutic applications.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Senlin Chai
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjin Yan
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Zhihong Xu
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Bin Zhao
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| |
Collapse
|
16
|
Savina IN, Zoughaib M, Yergeshov AA. Design and Assessment of Biodegradable Macroporous Cryogels as Advanced Tissue Engineering and Drug Carrying Materials. Gels 2021; 7:79. [PMID: 34203439 PMCID: PMC8293244 DOI: 10.3390/gels7030079] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Cryogels obtained by the cryotropic gelation process are macroporous hydrogels with a well-developed system of interconnected pores and shape memory. There have been significant recent advancements in our understanding of the cryotropic gelation process, and in the relationship between components, their structure and the application of the cryogels obtained. As cryogels are one of the most promising hydrogel-based biomaterials, and this field has been advancing rapidly, this review focuses on the design of biodegradable cryogels as advanced biomaterials for drug delivery and tissue engineering. The selection of a biodegradable polymer is key to the development of modern biomaterials that mimic the biological environment and the properties of artificial tissue, and are at the same time capable of being safely degraded/metabolized without any side effects. The range of biodegradable polymers utilized for cryogel formation is overviewed, including biopolymers, synthetic polymers, polymer blends, and composites. The paper discusses a cryotropic gelation method as a tool for synthesis of hydrogel materials with large, interconnected pores and mechanical, physical, chemical and biological properties, adapted for targeted biomedical applications. The effect of the composition, cross-linker, freezing conditions, and the nature of the polymer on the morphology, mechanical properties and biodegradation of cryogels is discussed. The biodegradation of cryogels and its dependence on their production and composition is overviewed. Selected representative biomedical applications demonstrate how cryogel-based materials have been used in drug delivery, tissue engineering, regenerative medicine, cancer research, and sensing.
Collapse
Affiliation(s)
- Irina N. Savina
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, UK
| | - Mohamed Zoughaib
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; (M.Z.); (A.A.Y.)
| | - Abdulla A. Yergeshov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; (M.Z.); (A.A.Y.)
| |
Collapse
|
17
|
Xing D, Liu W, Wang B, Li JJ, Zhao Y, Li H, Liu A, Du Y, Lin J. Intra-articular Injection of Cell-laden 3D Microcryogels Empower Low-dose Cell Therapy for Osteoarthritis in a Rat Model. Cell Transplant 2021; 29:963689720932142. [PMID: 32608995 PMCID: PMC7563831 DOI: 10.1177/0963689720932142] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Intra-articular injection of mesenchymal stem cells (MSCs) in an osteoarthritic joint can help slow down cartilage destruction. However, cell survival and the efficiency of repair are generally low due to mechanical damage during injection and a high rate of cell loss. We, thus, investigated an improved strategy for cell delivery to an osteoarthritic joint through the use of three-dimensional (3D) microcryogels. MSCs were seeded into 3D microcryogels. The viability and proliferation of MSCs in microcryogels were determined over 5 d, and the phenotype of MSCs was confirmed through trilineage differentiation tests and flow cytometry. In Sprague Dawley rats with induced osteoarthritis (OA) of the knee joint, a single injection was made with the following groups: saline control, low-dose free MSCs (1 × 105 cells), high-dose free MSCs (1 × 106 cells), and microcryogels + MSCs (1 × 105 cells). Cartilage degeneration was evaluated by macroscopic examination, micro-computed tomographic analysis, and histology. MSCs grown in microcryogels exhibited optimal viability and proliferation at 3 d with stable maintenance of phenotype in vitro. Microcryogels seeded with MSCs were, therefore, primed for 3 d before being used for in vivo experiments. At 4 and 8 wk, the microcryogels + MSCs and high-dose free MSC groups had significantly higher International Cartilage Repair Society macroscopic scores, histological evidence of more proteoglycan deposition and less cartilage loss accompanied by a lower Mankin score, and minimal radiographic evidence of osteoarthritic changes in the joint compared to the other two groups. In conclusion, intra-articular injection of cell-laden 3D microcryogels containing a low dose of MSCs can achieve similar effects as a high dose of free MSCs for OA in a rat model. Primed MSCs in 3D microcryogels can be considered as an improved delivery strategy for cell therapy in treating OA that minimizes cell dose while retaining therapeutic efficacy.
Collapse
Affiliation(s)
- Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, China.,Arthritis Institute, Peking University, Beijing, China.,These authors contributed equally to this article
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China.,Beijing Cytoniche Biotechnology Co, Ltd., Beijing, China.,These authors contributed equally to this article
| | - Bin Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.,These authors contributed equally to this article
| | - Jiao Jiao Li
- Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Yu Zhao
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, China.,Arthritis Institute, Peking University, Beijing, China
| | - Hui Li
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, China.,Arthritis Institute, Peking University, Beijing, China
| | - Aifeng Liu
- Department of Orthopedics, The First affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, China.,Arthritis Institute, Peking University, Beijing, China
| |
Collapse
|
18
|
Newland B, Newland H, Lorenzi F, Eigel D, Welzel PB, Fischer D, Wang W, Freudenberg U, Rosser A, Werner C. Injectable Glycosaminoglycan-Based Cryogels from Well-Defined Microscale Templates for Local Growth Factor Delivery. ACS Chem Neurosci 2021; 12:1178-1188. [PMID: 33754692 PMCID: PMC8033563 DOI: 10.1021/acschemneuro.1c00005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
![]()
Glycosaminoglycan-based hydrogels
hold great potential for applications
in tissue engineering and regenerative medicine. By mimicking the
natural extracellular matrix processes of growth factor binding and
release, such hydrogels can be used as a sustained delivery device
for growth factors. Since neural networks commonly follow well-defined,
high-aspect-ratio paths through the central and peripheral nervous
system, we sought to create a fiber-like, elongated growth factor
delivery system. Cryogels, with networks formed at subzero temperatures,
are well-suited for the creation of high-aspect-ratio biomaterials,
because they have a macroporous structure making them mechanically
robust (for ease of handling) yet soft and highly compressible (for
interfacing with brain tissue). Unlike hydrogels, cryogels can be
synthesized in advance of their use, stored with ease, and rehydrated
quickly to their original shape. Herein, we use solvent-assisted microcontact
molding to form sacrificial templates, in which we produced highly
porous cryogel microscale scaffolds with a well-defined elongated
shape via the photopolymerization of poly(ethylene glycol) diacrylate
and maleimide-functionalized heparin. Dissolution of the template
yielded cryogels that could load nerve growth factor (NGF) and release
it over a period of 2 weeks, causing neurite outgrowth in PC12 cell
cultures. This microscale template-assisted synthesis technique allows
tight control over the cryogel scaffold dimensions for high reproducibility
and ease of injection through fine gauge needles.
Collapse
Affiliation(s)
- Ben Newland
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, U.K
| | - Heike Newland
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Francesca Lorenzi
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Francesco Marzolo, 135131 Padova, Italy
| | - Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Petra B. Welzel
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Dieter Fischer
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Wenxin Wang
- Charles Institute for Dermatology, University College Dublin, Dublin D04 V1W8, Ireland
| | - Uwe Freudenberg
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Anne Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff CF10 3AX, U.K
- Brain Repair And Intracranial Neurotherapeutics (BRAIN) Unit, Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis
Building, Maindy Road, Cardiff CF24 4HQ3, U.K
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
19
|
Çimen D, Özbek MA, Bereli N, Mattiasson B, Denizli A. Injectable Cryogels in Biomedicine. Gels 2021; 7:gels7020038. [PMID: 33915687 PMCID: PMC8167568 DOI: 10.3390/gels7020038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
Cryogels are interconnected macroporous materials that are synthesized from a monomer solution at sub-zero temperatures. Cryogels, which are used in various applications in many research areas, are frequently used in biomedicine applications due to their excellent properties, such as biocompatibility, physical resistance and sensitivity. Cryogels can also be prepared in powder, column, bead, sphere, membrane, monolithic, and injectable forms. In this review, various examples of recent developments in biomedical applications of injectable cryogels, which are currently scarce in the literature, made from synthetic and natural polymers are discussed. In the present review, several biomedical applications of injectable cryogels, such as tissue engineering, drug delivery, therapeutic, therapy, cell transplantation, and immunotherapy, are emphasized. Moreover, it aims to provide a different perspective on the studies to be conducted on injectable cryogels, which are newly emerging trend.
Collapse
Affiliation(s)
- Duygu Çimen
- Department of Chemistry, Hacettepe University, Ankara 06800, Turkey; (D.Ç.); (M.A.Ö.); (N.B.)
| | - Merve Asena Özbek
- Department of Chemistry, Hacettepe University, Ankara 06800, Turkey; (D.Ç.); (M.A.Ö.); (N.B.)
| | - Nilay Bereli
- Department of Chemistry, Hacettepe University, Ankara 06800, Turkey; (D.Ç.); (M.A.Ö.); (N.B.)
| | - Bo Mattiasson
- Department of Biotechnology, Lund University, Box 124, 221 00 Lund, Sweden;
| | - Adil Denizli
- Department of Chemistry, Hacettepe University, Ankara 06800, Turkey; (D.Ç.); (M.A.Ö.); (N.B.)
- Correspondence:
| |
Collapse
|
20
|
Marsico G, Martin‐Saldaña S, Pandit A. Therapeutic Biomaterial Approaches to Alleviate Chronic Limb Threatening Ischemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003119. [PMID: 33854887 PMCID: PMC8025020 DOI: 10.1002/advs.202003119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/24/2020] [Indexed: 05/14/2023]
Abstract
Chronic limb threatening ischemia (CLTI) is a severe condition defined by the blockage of arteries in the lower extremities that leads to the degeneration of blood vessels and is characterized by the formation of non-healing ulcers and necrosis. The gold standard therapies such as bypass and endovascular surgery aim at the removal of the blockage. These therapies are not suitable for the so-called "no option patients" which present multiple artery occlusions with a likelihood of significant limb amputation. Therefore, CLTI represents a significant clinical challenge, and the efforts of developing new treatments have been focused on stimulating angiogenesis in the ischemic muscle. The delivery of pro-angiogenic nucleic acid, protein, and stem cell-based interventions have limited efficacy due to their short survival. Engineered biomaterials have emerged as a promising method to improve the effectiveness of these latter strategies. Several synthetic and natural biomaterials are tested in different formulations aiming to incorporate nucleic acid, proteins, stem cells, macrophages, or endothelial cells in supportive matrices. In this review, an overview of the biomaterials used alone and in combination with growth factors, nucleic acid, and cells in preclinical models is provided and their potential to induce revascularization and regeneration for CLTI applications is discussed.
Collapse
Affiliation(s)
- Grazia Marsico
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Sergio Martin‐Saldaña
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| |
Collapse
|
21
|
Eigel D, Werner C, Newland B. Cryogel biomaterials for neuroscience applications. Neurochem Int 2021; 147:105012. [PMID: 33731275 DOI: 10.1016/j.neuint.2021.105012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials in the form of 3D polymeric scaffolds have been used to create structurally and functionally biomimetic constructs of nervous system tissue. Such constructs can be used to model defects and disease or can be used to supplement neuronal tissue regeneration and repair. One such group of biomaterial scaffolds are hydrogels, which have been widely investigated for cell/tissue culture and as cell or molecule delivery systems in the field of neurosciences. However, a subset of hydrogels called cryogels, have shown to possess several distinct structural advantages over conventional hydrogel networks. Their macroporous structure, created via the time and resource efficient fabrication process (cryogelation) not only allows mass fluid transport throughout the structure, but also creates a high surface area to volume ratio for cell growth or drug loading. In addition, the macroporous structure of cryogels is ideal for applications in the central nervous system as they are very soft and spongey, yet also robust, which makes them a user-friendly and reproducible tool to address neuroscience challenges. In this review, we aim to provide the neuroscience community, who may not be familiar with the fundamental concepts of cryogels, an accessible summary of the basic information that pertain to their use in the brain and nervous tissue. We hope that this review shall initiate creative ways that cryogels could be further adapted and employed to tackle unsolved neuroscience challenges.
Collapse
Affiliation(s)
- Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany; Technische Universität Dresden, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ben Newland
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany; School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF10 3NB, Cardiff, Wales, UK.
| |
Collapse
|
22
|
Yuan Z, Yuan X, Zhao Y, Cai Q, Wang Y, Luo R, Yu S, Wang Y, Han J, Ge L, Huang J, Xiong C. Injectable GelMA Cryogel Microspheres for Modularized Cell Delivery and Potential Vascularized Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006596. [PMID: 33620759 DOI: 10.1002/smll.202006596] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/03/2021] [Indexed: 06/12/2023]
Abstract
Cell therapeutics hold tremendous regenerative potential and the therapeutic effect depends on the effective delivery of cells. However, current cell delivery carriers with unsuitable cytocompatibility and topological structure demonstrate poor cell viability during injection. Therefore, porous shape-memory cryogel microspheres (CMS) are prepared from methacrylated gelatin (GelMA) by combining an emulsion technique with gradient-cooling cryogelation. Pore sizes of the CMS are adjusted via the gradient-cooling procedure, with the optimized pore size (15.5 ± 6.0 µm) being achieved on the 30-min gradient-cooled variant (CMS-30). Unlike hydrogel microspheres (HMS), CMS promotes human bone marrow stromal cell (hBMSC) and human umbilical vein endothelial cell (HUVEC) adhesion, proliferated with high levels of stemness for 7 d, and protects cells during the injection process using a 26G syringe needle. Moreover, CMS-30 enhances the osteogenic differentiation of hBMSCs in osteoinductive media. CMS can serve as building blocks for delivering multiple cell types. Here, hBMSC-loaded and HUVEC-loaded CMS-30, mixed at a 1:1 ratio, are injected subcutaneously into nude mice for 2 months. Results show the development of vascularized bone-like tissue with high levels of OCN and CD31. These findings indicate that GelMA CMS of a certain pore size can effectively deliver multiple cells to achieve functional tissue regeneration.
Collapse
Affiliation(s)
- Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Xiaojing Yuan
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Yuming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yue Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Ruochen Luo
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Shi Yu
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Yuanyuan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Jianmin Han
- Dental Medical Devices Testing Center, Dental Materials Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Lihong Ge
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| |
Collapse
|
23
|
Kaur S, Kaur I, Rawal P, Tripathi DM, Vasudevan A. Non-matrigel scaffolds for organoid cultures. Cancer Lett 2021; 504:58-66. [PMID: 33582211 DOI: 10.1016/j.canlet.2021.01.025] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/19/2022]
Abstract
Organoids are three-dimensional cell cultures mostly from tissue-resident or embryonic stem cells (one or multiple) on hydrogels along with defined growth factors. Currently, matrigel is the most commonly employed matrix for 3D organoid cultures. However, certain undesirable attributes of matrigel have paved the way for several other natural and synthetic hydrogel scaffolds for organoid cultures. In this review, we discuss the constraints of matrigel and describe other alternative scaffolds that have been used for organoid cultures. Given the potential of organoids in a plethora of therapeutic and pharmaceutical applications, it is indeed imperative to develop defined and customized hydrogels other than the matrigel.
Collapse
Affiliation(s)
- Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Preety Rawal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India; School of Biotechnology, Gautam Buddha University, Greater Noida, UP, India
| | - Dinesh M Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
24
|
Shiekh PA, Andrabi SM, Singh A, Majumder S, Kumar A. Designing cryogels through cryostructuring of polymeric matrices for biomedical applications. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Çetin K, Aslıyüce S, Idil N, Denizli A. Preparation of lysozyme loaded gelatin microcryogels and investigation of their antibacterial properties. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:189-204. [PMID: 32962559 DOI: 10.1080/09205063.2020.1825303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Antibacterial micron-sized cryogels, so-called microcryogels, were prepared by cryogelation of gelatin and integration of lysozyme. Gelation yield, specific surface area, macro-porosity and swelling degree of the microcryogels were examined in order to characterize their physical properties. MTT method was utilized to measure cell viability of the gelatin microcryogels with a period of 24, 48, and 72 h and no significant decrease was observed at 72 h. Apoptotic staining assay also showed high viability at 24, 48, 72 h in parallel with the control group. The antibacterial performances of the gelatin microcryogels against Bacillus subtilis, Staphylococcus aureus, and Escherichia coli were examined. The results showed that the incorporation of lysozyme into gelatin microcryogels exhibited the antibacterial activity against S. aureus, B. subtilis, and E. coli, that may provide great potential for various applications in the biomedical industry.
Collapse
Affiliation(s)
- Kemal Çetin
- Department of Biomedical Engineering, Necmettin Erbakan University, Konya, Turkey
| | - Sevgi Aslıyüce
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Neslihan Idil
- Department of Biology, Hacettepe University, Ankara, Turkey
| | - Adil Denizli
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
26
|
Yang X, Meng H, Peng J, Xu L, Wang Y, Sun X, Zhao Y, Quan Q, Yu W, Chen M, Shi T, Du Y, Lu S, Wang A. Construction of Microunits by Adipose-Derived Mesenchymal Stem Cells Laden with Porous Microcryogels for Repairing an Acute Achilles Tendon Rupture in a Rat Model. Int J Nanomedicine 2020; 15:7155-7171. [PMID: 33061373 PMCID: PMC7535144 DOI: 10.2147/ijn.s238399] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 08/28/2020] [Indexed: 01/07/2023] Open
Abstract
Objective Tissue engineering approaches seem to be an attractive therapy for tendon rupture. Novel injectable porous gelatin microcryogels (GMs) can promote cell attachment and proliferation, thus facilitating the repair potential for target tissue regeneration. The research objectives of this study were to assess the efficacy of tissue-like microunits constructed by multiple GMs laden with adipose-derived mesenchymal stem cells (ASCs) in accelerated tendon regeneration in a rat model. Methods Through a series of experiments, such as isolation and identification of ASCs, scanning electron microscopy, mercury intrusion porosimetry (MIP), laser scanning confocal microscopy and the CCK-8 test, the biocompatibility of GMs was evaluated. In an in vivo study, 64 rat right transected Achilles tendons were randomly divided into four groups: the ASCs+GMs group (microunits aggregated by multiple ASC-laden GMs injected into the gap), the ASCs group (ASCs injected into the gap), the GMs group (GMs injected into the gap) and the blank defect group (non-treated). At 2 and 4 weeks postoperatively, the healing tissue was harvested to evaluate the gross observation and scoring, biomechanical testing, histological staining and quantitative scoring. Gait analysis was performed over time. The 64 rats were randomly assigned into 4 groups: (1) micro-unit group (ASCs+GMs) containing ASC (105)-loaded 120 GMs in 60 μL DMEM; (2) cell control group (ASCs) containing 106 ASCs in 60 μL DMEM; (3) GM control group (GMs) containing 120 blank GMs in 60 μL DMEM; (4) blank defect group (Defect) containing 60 μL DMEM, which were injected into the defect sites. All animals were sacrificed at 2 and 4 weeks postsurgery (Table 1). Results In an in vitro study, GMs (from 126 μm to 348 μm) showed good porosities and a three-dimensional void structure with a good interpore connectivity of the micropores and exhibited excellent biocompatibility with ASCs. As the culture time elapsed, the extracellular matrix (ECM) secreted by ASCs encased the GMs, bound multiple microspheres together, and then formed active tendon tissue-engineering microunits. In animal experiments, the ASCs+GMs group and the ASCs group showed stimulatory effects on Achilles tendon healing. Moreover, the ASCs+GMs group was the best at improving the macroscopic appearance, histological morphology, Achilles functional index (AFI), and biomechanical properties of repair tissue without causing adverse immune reactions. Conclusion Porous GMs were conducive to promoting cell proliferation and facilitating ECM secretion. The ASCs-GMs matrices showed an obvious therapeutic efficiency for Achilles tendon rupture in rats.
Collapse
Affiliation(s)
- Xuan Yang
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China.,Department of orthopedics, The First Hospital of Shanxi Medical University YanHu District Branch, Yuncheng, Shanxi 044000, People's Republic of China
| | - Haoye Meng
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Jiang Peng
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Lijuan Xu
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Yu Wang
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Xun Sun
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, People's Republic of China
| | - Yanxu Zhao
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Qi Quan
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Wen Yu
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Mingxue Chen
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Tong Shi
- Department of orthopedics, The First Hospital of Shanxi Medical University YanHu District Branch, Yuncheng, Shanxi 044000, People's Republic of China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Shibi Lu
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Aiyuan Wang
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &war Injuries PLA, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| |
Collapse
|
27
|
Xing D, Liu W, Li JJ, Liu L, Guo A, Wang B, Yu H, Zhao Y, Chen Y, You Z, Lyu C, Li W, Liu A, Du Y, Lin J. Engineering 3D functional tissue constructs using self-assembling cell-laden microniches. Acta Biomater 2020; 114:170-182. [PMID: 32771588 DOI: 10.1016/j.actbio.2020.07.058] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
Tissue engineering using traditional size fixed scaffolds and injectable biomaterials are faced with many limitations due to the difficulties of producing macroscopic functional tissues. In this study, 3D functional tissue constructs were developed by inducing self-assembly of microniches, which were cell-laden gelatin microcryogels. During self-assembly, the accumulation of extracellular matrix (ECM) components was found to strengthen cell-cell and cell-ECM interactions, leading to the construction of a 'native' microenvironment that better preserved cell viability and functions. MSCs grown in self-assembled constructs showed increased maintenance of stemness, reduced senescence and improved paracrine activity compared with cells grown in individual microniches without self-assembly. As an example of applying the self-assembled constructs in tissue regeneration, the constructs were used to induce in vivo articular cartilage repair and successfully regenerated hyaline-like cartilage tissue in the absence of other extrinsic factors. This unique approach of developing self-assembled 3D functional constructs holds great promise for the generation of tissue engineered organoids and repair of challenging tissue defects. STATEMENT OF SIGNIFICANCE: We developed 3D functional tissue constructs using a unique gelatin-based microscopic hydrogel (microcryogels). Mesenchymal stem cells (MSCs) were loaded into gelatin microcryogels to form microscopic cell-laden units (microniches), which were induced to undergo self-assembly using a specially designed 3D printed frame. Extracellular matrix accumulation among the microniches resulted in self-assembled macroscopic constructs with superior ability to maintain the phenotypic characteristics and stemness of MSCs, together with the suppression of senescence and enhanced paracrine function. As an example of application in tissue regeneration, the self-assembled constructs were shown to successfully repair articular cartilage defects without any other supplements. This unique strategy for developing 3D functional tissue constructs allows the optimisation of stem cell functions and construction of biomimetic tissue organoids.
Collapse
Affiliation(s)
- Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China; Arthritis Institute, Peking University, Beijing 100044, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney (UTS), Ultimo, NSW 2007, Australia
| | - Longwei Liu
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anqi Guo
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210009, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yu Zhao
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China; Arthritis Institute, Peking University, Beijing 100044, China
| | - Yuling Chen
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Cheng Lyu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Aifeng Liu
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China.
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China; Arthritis Institute, Peking University, Beijing 100044, China.
| |
Collapse
|
28
|
Wu J, Li G, Ye T, Lu G, Li R, Deng L, Wang L, Cai M, Cui W. Stem cell-laden injectable hydrogel microspheres for cancellous bone regeneration. CHEMICAL ENGINEERING JOURNAL 2020; 393:124715. [DOI: 10.1016/j.cej.2020.124715] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
29
|
Newland B, Varricchio C, Körner Y, Hoppe F, Taplan C, Newland H, Eigel D, Tornillo G, Pette D, Brancale A, Welzel PB, Seib FP, Werner C. Focal drug administration via heparin-containing cryogel microcarriers reduces cancer growth and metastasis. Carbohydr Polym 2020; 245:116504. [PMID: 32718615 DOI: 10.1016/j.carbpol.2020.116504] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/11/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Developing drug delivery systems that release anticancer drugs in a controlled and sustained manner remains challenging. We hypothesized that highly sulfated heparin-based microcarriers would allow electrostatic drug binding and controlled release. In silico modelling showed that the anticancer drug doxorubicin has affinity for the heparin component of the microcarriers. Experimental results showed that the strong electrostatic interaction was reversible, allowing both doxorubicin loading and a subsequent slow release over 42 days without an initial burst release. The drug-loaded microcarriers were able to reduce cancer cell viability in vitro in both hormone-dependent and highly aggressive triple-negative human breast cancer cells. Focal drug treatment, of an in vivo orthotopic triple-negative breast cancer model significantly decreased tumor burden and reduced cancer metastasis, whereas systemic administration of an equivalent drug dose was ineffective. This study proves that heparin-based microcarriers can be used as drug delivery platforms, for focal delivery and sustained long-term drug release.
Collapse
Affiliation(s)
- Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK; Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany.
| | - Carmine Varricchio
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Yvonne Körner
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - Franziska Hoppe
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - Christian Taplan
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - Heike Newland
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - Giusy Tornillo
- European Cancer Stem Cell Research Institute, School of Biosciences, Hadyn Ellis Building, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Dagmar Pette
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Petra B Welzel
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany
| | - F Philipp Seib
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK; EPSRC Future Manufacturing Research Hub for Continuous Manufacturing and Advanced Crystallisation (CMAC), University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069, Dresden, Germany; Technische Universität Dresden, Center for Regenerative Therapies Dresden, Fetscherstr. 105, 01307, Dresden, Germany
| |
Collapse
|
30
|
Eggermont LJ, Rogers ZJ, Colombani T, Memic A, Bencherif SA. Injectable Cryogels for Biomedical Applications. Trends Biotechnol 2020; 38:418-431. [DOI: 10.1016/j.tibtech.2019.09.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022]
|
31
|
Mizuno Y, Taguchi T. A hydrophobic gelatin fiber sheet promotes secretion of endogenous vascular endothelial growth factor and stimulates angiogenesis. RSC Adv 2020; 10:24800-24807. [PMID: 35517459 PMCID: PMC9055140 DOI: 10.1039/d0ra03593a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/20/2020] [Indexed: 01/24/2023] Open
Abstract
In tissue engineering and regenerative medicine, the formation of vascular beds is an effective method to supply oxygen and nutrients to implanted cells or tissues to improve their survival and promote normal cellular functions. Various types of angiogenic materials have been developed by incorporating growth factors, such as vascular endothelial growth factor, in biocompatible materials. However, these exogenous growth factors suffer from instability and inactivation under physiological conditions. In this study, we designed a novel angiogenic electrospun fiber sheet (C16-FS) composed of Alaska pollock-derived gelatin (ApGltn) modified with hexadecyl (C16) groups to induce localized and sustained angiogenesis without growth factors. C16-FS was thermally crosslinked to enhance its stability. We demonstrated that C16-FS swells in phosphate-buffered saline for over 24 h and resists degradation. Laser doppler perfusion imaging showed that C16-FS induced increased blood perfusion when implanted subcutaneously in rats compared with unmodified ApGltn-fiber sheets (Org-FS) and the sham control. Furthermore, angiogenesis was sustained for up to 7 days following implantation. Immunohistochemical studies revealed elevated nuclear factor-κB and CD31 levels around the C16-FS implantation site compared with the Org-FS implantation site and the control incision site. These results demonstrate that C16-FS is a promising angiogenic material to promote the formation of vascular beds for cell and tissue transplantation without the need for growth factors. In vivo long-term growth factor-free angiogenesis by LPS-mimicking C16-modified gelatin based electrospun fiber sheet.![]()
Collapse
Affiliation(s)
- Yosuke Mizuno
- Graduate School of Science and Technology
- University of Tsukuba
- Tsukuba
- Japan
- Polymers and Biomaterials Field
| | - Tetsushi Taguchi
- Graduate School of Science and Technology
- University of Tsukuba
- Tsukuba
- Japan
- Polymers and Biomaterials Field
| |
Collapse
|
32
|
Kumari J, Teotia AK, Karande AA, Kumar A. A minimally-invasive cryogel based approach for the development of human ectopic liver in a mouse model. J Biomed Mater Res B Appl Biomater 2019; 108:1022-1032. [PMID: 31397074 DOI: 10.1002/jbm.b.34454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/10/2019] [Accepted: 07/09/2019] [Indexed: 11/09/2022]
Abstract
Human liver tissue is preferable over nonhuman liver tissue for preclinical drug screening, as the former can better predict side effects specific to humans. However, due to limited supply and ethical issues with human liver tissue, it is desirable to develop an animal model having functional human liver tissue. In this study, we have established an ectopic functional human liver tissue in a mouse model, using a minimally-invasive method. Firstly, a human liver tissue mass using HepG2 cells and poly(N-isopropylacrylamide) (PNIPAAm) incorporated poly(ethylene glycol)-alginate-gelatin (PAG) cryogel matrix was developed in vitro. It was later implanted in mouse peritoneal cavity using a 16 G needle. Viscoelastic nature along with low Young's modulus provided injectable properties to the cryogel. We confirmed minimal cell loss/death while injecting. Further, by in vivo study efficacy of both injectable and surgical implantation approaches were compared. No significant difference in terms of cell infiltration, human serum albumin (HSA) secretion and enzyme activity confirmed efficacy. This model developed using a minimally-invasive approach can overcome the limitations of surgical implantation due to its cost effective and user friendly nature.
Collapse
Affiliation(s)
- Jyoti Kumari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP, India
| | - Arun K Teotia
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP, India
| | - Anjali A Karande
- Department of Biochemistry, Indian Institute of Sciences, Bangalore, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, UP, India
| |
Collapse
|
33
|
Mizuno Y, Taguchi T. Promotion of Cell Migration into a Hydrophobically modified Alaska Pollock Gelatin‐Based Hydrogel. Macromol Biosci 2019; 19:e1900083. [DOI: 10.1002/mabi.201900083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/26/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Yosuke Mizuno
- Graduate School of Pure and Applied SciencesUniversity of Tsukuba 1–1–1 Tennodai Tsukuba Ibaraki 305–8577 Japan
| | - Tetsushi Taguchi
- Graduate School of Pure and Applied SciencesUniversity of Tsukuba 1–1–1 Tennodai Tsukuba Ibaraki 305–8577 Japan
- Biomaterials FieldResearch Center for Functional MaterialsNational Institute for Materials Science 1–1 Namiki Tsukuba Ibaraki 305–0044 Japan
| |
Collapse
|
34
|
Çetin K, Denizli A. Immunoaffinity microcryogels for purification of transferrin. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1114-1115:5-12. [DOI: 10.1016/j.jchromb.2019.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/03/2019] [Accepted: 03/17/2019] [Indexed: 12/14/2022]
|
35
|
|
36
|
Memic A, Colombani T, Eggermont LJ, Rezaeeyazdi M, Steingold J, Rogers ZJ, Navare KJ, Mohammed HS, Bencherif SA. Latest Advances in Cryogel Technology for Biomedical Applications. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800114] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Adnan Memic
- Center of NanotechnologyKing Abdulaziz University Jeddah 21589 Saudi Arabia
- Center for Biomedical EngineeringDepartment of MedicineBrigham and Women's HospitalHarvard Medical School Cambridge MA 02139 USA
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
| | - Thibault Colombani
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
| | - Loek J. Eggermont
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
- Department of Tumor ImmunologyOncode Institute, Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen 6500 The Netherlands
| | | | - Joseph Steingold
- Department of Pharmaceutical SciencesNortheastern University Boston MA 02115 USA
| | - Zach J. Rogers
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
| | | | | | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
- Department of BioengineeringNortheastern University Boston MA 02115 USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard University Cambridge MA 02138 USA
- Sorbonne UniversityUTC CNRS UMR 7338Biomechanics and Bioengineering (BMBI)University of Technology of Compiègne Compiègne 60159 France
| |
Collapse
|
37
|
Kim I, Lee SS, Bae S, Lee H, Hwang NS. Heparin Functionalized Injectable Cryogel with Rapid Shape-Recovery Property for Neovascularization. Biomacromolecules 2018; 19:2257-2269. [PMID: 29689163 DOI: 10.1021/acs.biomac.8b00331] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cryogel based scaffolds have high porosity with interconnected macropores that may provide cell compatible microenvironment. In addition, cryogel based scaffolds can be utilized in minimally invasive surgery due to its sponge-like properties, including rapid shape recovery and injectability. Herein, we developed an injectable cryogel by conjugating heparin to gelatin as a carrier for vascular endothelial growth factor (VEGF) and fibroblasts in hindlimb ischemic disease. Our gelatin/heparin cryogel showed gelatin concentration-dependent mechanical properties, swelling ratios, interconnected porosities, and elasticities. In addition, controlled release of VEGF led to effective angiogenic responses both in vitro and in vivo. Furthermore, its sponge-like properties enabled cryogels to be applied as an injectable carrier system for in vivo cells and growth factor delivery. Our heparin functionalized injectable cryogel facilitated the angiogenic potential by facilitating neovascularization in a hindlimb ischemia model.
Collapse
Affiliation(s)
- Inseon Kim
- School of Chemical and Biological Engineering, the Institute of Chemical Processes , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Seunghun S Lee
- Interdisciplinary Program in Bioengineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Sunghoon Bae
- School of Chemical and Biological Engineering, the Institute of Chemical Processes , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Hoyon Lee
- School of Chemical and Biological Engineering, the Institute of Chemical Processes , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, the Institute of Chemical Processes , Seoul National University , Seoul , 08826 , Republic of Korea.,Interdisciplinary Program in Bioengineering , Seoul National University , Seoul , 08826 , Republic of Korea.,BioMAX/N-Bio Institute , Seoul National University , Seoul , 08826 , Republic of Korea
| |
Collapse
|
38
|
Jiang S, Li SC, Huang C, Chan BP, Du Y. Physical Properties of Implanted Porous Bioscaffolds Regulate Skin Repair: Focusing on Mechanical and Structural Features. Adv Healthc Mater 2018; 7:e1700894. [PMID: 29334185 DOI: 10.1002/adhm.201700894] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/23/2017] [Indexed: 01/07/2023]
Abstract
Porous bioscaffolds are applied to facilitate skin repair since the early 1990s, but a perfect regeneration outcome has yet to be achieved. Until now, most efforts have focused on modulating the chemical properties of bioscaffolds, while physical properties are traditionally overlooked. Recent advances in mechanobiology and mechanotherapy have highlighted the importance of biomaterials' physical properties in the regulation of cellular behaviors and regenerative processes. In skin repair, the mechanical and structural features of porous bioscaffolds are two major physical properties that determine therapeutic efficacy. Here, first an overview of natural skin repair with an emphasis on the major biophysically sensitive cell types involved in this multistage process is provided, followed by an introduction of the four roles of bioscaffolds as skin implants. Then, how the mechanical and structural features of bioscaffolds influence these four roles is discussed. The mechanical and structural features of porous bioscaffolds should be tailored to balance the acceleration of wound closure and functional improvements of the repaired skin. This study emphasizes that decoupling and precise control of the mechanical and structural features of bioscaffolds are significant aspects that should be considered in future biomaterial optimization, which can build a foundation to ultimately achieve perfect skin regeneration outcomes.
Collapse
Affiliation(s)
- Shumeng Jiang
- Department of Biomedical Engineering School of Medicine Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| | - Sabrina Cloud Li
- Department of Biomedical Engineering School of Medicine Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| | - Chenyu Huang
- Beijing Tsinghua Changgung Hospital Tsinghua University Beijing 102218 China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory Department of Mechanical Engineering The University of Hong Kong Hong Kong Special Administrative Region China
| | - Yanan Du
- Department of Biomedical Engineering School of Medicine Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| |
Collapse
|
39
|
Injectable nanohydroxyapatite-chitosan-gelatin micro-scaffolds induce regeneration of knee subchondral bone lesions. Sci Rep 2017; 7:16709. [PMID: 29196647 PMCID: PMC5711958 DOI: 10.1038/s41598-017-17025-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023] Open
Abstract
Subchondral bone has been identified as an attractive target for KOA. To determine whether a minimally invasive micro-scaffolds could be used to induce regeneration of knee subchondral bone lesions, and to examine the protective effect of subchondral bone regeneration on upper cartilage, a ready-to-use injectable treatment with nanohydroxyapatite-chitosan-gelatin micro-scaffolds (HaCGMs) is proposed. Human-infrapatellar-fat-pad-derived adipose stem cells (IPFP-ASCs) were used as a cellular model to examine the osteo-inductivity and biocompatibility of HaCGMs, which were feasibly obtained with potency for multi-potential differentiations. Furthermore, a subchondral bone lesion model was developed to mimic the necrotic region removing performed by surgeons before sequestrectomy. HaCGMs were injected into the model to induce regeneration of subchondral bone. HaCGMs exhibited desirable swelling ratios, porosity, stiffness, and bioactivity and allowed cellular infiltration. Eight weeks after treatment, assessment via X-ray imaging, micro-CT imaging, and histological analysis revealed that rabbits treated with HaCGMs had better subchondral bone regeneration than those not treated. Interestingly, rabbits in the HaCGM treatment group also exhibited improved reservation of upper cartilage compared to those in other groups, as shown by safranin O-fast green staining. Present study provides an in-depth demonstration of injectable HaCGM-based regenerative therapy, which may provide an attractive alternative strategy for treating KOA.
Collapse
|
40
|
Xia P, Zhang K, Gong Y, Li G, Yan S, Yin J. Injectable Stem Cell Laden Open Porous Microgels That Favor Adipogenesis: In Vitro and in Vivo Evaluation. ACS APPLIED MATERIALS & INTERFACES 2017; 9:34751-34761. [PMID: 28930432 DOI: 10.1021/acsami.7b13065] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Microgels, with large surface area per volume, show great advantages in adipose tissue engineering due to their injectability and similarity with natural extracellular matrix. However, to date, no studies have tried applying microgels to adipose tissue regeneration. Herein, based on double-bonded poly(l-glutamic acid)-g-2-hydroxyethyl methacrylate (PLGA-g-HEMA) and maleic anhydride-modified chitosan (MCS), an open porous microgel with high hydrophilicity and great injectability is successfully prepared (microgels diameters of 200-300 μm, pore diameter of 38 μm, and porosity of 88.3%). The storage modulus of 30 mg/mL of the microgel dispersions is 2000 Pa, which is similar to that of the native adipose tissue. The spheroidal stem cell shape and extensive cell-cell connections can be formed in the present microgels to promote adipogenic differentiation and realize adipose tissue regeneration. After injection in vitro, the microgels can maintain high stem cell viability up to 14 days. The extensive Oil red O staining is observed after adipogenic induction for 14 days. After 12 weeks postimplantation, adipose tissues can be regenerated well. Blood vessels are formed in the neogenerated tissues. The degradation rate of microgels roughly matches with the adipose tissue formation rate. The study offers an applicable microgel system to boost the adipose tissue regeneration.
Collapse
Affiliation(s)
- Pengfei Xia
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Kunxi Zhang
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Yan Gong
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Guifei Li
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Shifeng Yan
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| | - Jingbo Yin
- Department of Polymer Materials, Shanghai University , 99 Shangda Road, Shanghai 200444, People's Republic of China
| |
Collapse
|
41
|
Li Y, Yan X, Liu W, Zhou L, You Z, Du Y. 3D Microtissues for Injectable Regenerative Therapy and High-throughput Drug Screening. J Vis Exp 2017:55982. [PMID: 29053690 PMCID: PMC5752368 DOI: 10.3791/55982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
To upgrade traditional 2D cell culture to 3D cell culture, we have integrated microfabrication with cryogelation technology to produce macroporous microscale cryogels (microcryogels), which can be loaded with a variety of cell types to form 3D microtissues. Herein, we present the protocol to fabricate versatile 3D microtissues and their applications in regenerative therapy and drug screening. Size and shape-controllable microcryogels can be fabricated on an array chip, which can be harvested off-chip as individual cell-loaded carriers for injectable regenerative therapy or be further assembled on-chip into 3D microtissue arrays for high-throughput drug screening. Due to the high elastic nature of these microscale cryogels, the 3D microtissues exhibit great injectability for minimally invasive cell therapy by protecting cells from mechanical shear force during injection. This ensures enhanced cell survival and therapeutic effect in the mouse limb ischemia model. Meanwhile, assembly of 3D microtissue arrays in a standard 384-multi-well format facilitates the use of common laboratory facilities and equipment, enabling high-throughput drug screening on this versatile 3D cell culture platform.
Collapse
Affiliation(s)
- Yaqian Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua University; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases
| | - Xiaojun Yan
- Department of Biomedical Engineering, School of Medicine, Tsinghua University
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University
| | - Lyu Zhou
- Department of Biomedical Engineering, School of Medicine, Tsinghua University; School of Life Sciences, Tsinghua University
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua University
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua University; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases;
| |
Collapse
|
42
|
Salehi S, Ostrovidov S, Ebrahimi M, Sadeghian RB, Liang X, Nakajima K, Bae H, Fujie T, Khademhosseini A. Development of Flexible Cell-Loaded Ultrathin Ribbons for Minimally Invasive Delivery of Skeletal Muscle Cells. ACS Biomater Sci Eng 2017; 3:579-589. [PMID: 33429625 DOI: 10.1021/acsbiomaterials.6b00696] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell transplantation therapy provides a potential solution for treating skeletal muscle disorders, but cell survival after transplantation is poor. This limitation could be addressed by grafting donor cells onto biomaterials to protect them against harsh environments and processing, consequently improving cell viability in situ. Thus, we present here the fabrication of poly(lactic-co-glycolic acid) (PLGA) ultrathin ribbons with "canal-like" structures using a microfabrication technique to generate ribbons of aligned murine skeletal myoblasts (C2C12). We found that the ribbons functionalized with a solution of 3,4-dihydroxy-l-phenylalanine (DOPA) and then coated with poly-l-lysine (PLL) and fibronectin (FN) improve cell attachment and support the growth of C2C12. The viability of cells on the ribbons is evaluated following the syringe-handling steps of injection with different needle sizes. C2C12 cells readily adhere to the ribbon surface, proliferate over time, align (over 74%), maintain high viability (over 80%), and differentiate to myotubes longer than 400 μm. DNA content quantification carried out before and after injection and myogenesis evaluation confirm that cell-loaded ribbons can safely retain cells with high functionality after injection and are suitable for minimally invasive cell transplantation.
Collapse
Affiliation(s)
- Sahar Salehi
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Serge Ostrovidov
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Majid Ebrahimi
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Ramin Banan Sadeghian
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Xiaobin Liang
- School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| | - Ken Nakajima
- School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| | - Hojae Bae
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul 143-701, Republic of Korea
| | - Toshinori Fujie
- Waseda Institute for Advanced Study, Waseda University, Shinjuku, Tokyo 162-8480, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Ali Khademhosseini
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan.,Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul 143-701, Republic of Korea.,Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02139, United States.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States.,Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
43
|
Qi C, Li Y, Badger P, Yu H, You Z, Yan X, Liu W, Shi Y, Xia T, Dong J, Huang C, Du Y. Pathology-targeted cell delivery via injectable micro-scaffold capsule mediated by endogenous TGase. Biomaterials 2017; 126:1-9. [PMID: 28237907 DOI: 10.1016/j.biomaterials.2017.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 12/13/2022]
Abstract
Targeted cell delivery to lesion sites via minimally invasive approach remains an unmet need in regenerative medicine to endow satisfactory therapeutic efficacy and minimized side-effects. Here, we rationally designed a pathology-targeted cell delivery strategy leveraging injectable micro-scaffolds as cell-loading capsule and endogenous tissue transglutaminase (TGase) at lesion site as adhesive. Up-regulated TGase post-liver injury catalyzed chemical bonding between the glutamine and lysine residues on liver surface and micro-scaffolds both ex vivo and in vivo, facilitating sufficient adhesion on the pathological liver. Upon intraperitoneal injection, Mesenchymal Stem Cell-loaded capsules, exhibiting cell protection from shear-induced damage and post-transplantation anoikis, adhered to the CCl4-treated liver with a hundred-fold improvement in targeting efficiency (70.72%) compared to free-cell injection, which dramatically improved mice survival (33.3% vs. 0% for free-cell therapy) even with low-dosage treatment. This unique and widely-applicable cell delivery mechanism and strategy hold great promise for transforming cell therapy for refractory diseases.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yaqian Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Patrick Badger
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaojun Yan
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yan Shi
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Tie Xia
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jiahong Dong
- Department of Hepatobiliary Surgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Chenyu Huang
- Department of Plastic, Reconstructive and Aesthetic Surgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou 310003, China.
| |
Collapse
|
44
|
Wang J, Chen F, Liu L, Qi C, Wang B, Yan X, Huang C, Hou W, Zhang MQ, Chen Y, Du Y. Engineering EMT using 3D micro-scaffold to promote hepatic functions for drug hepatotoxicity evaluation. Biomaterials 2016; 91:11-22. [PMID: 26994875 DOI: 10.1016/j.biomaterials.2016.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 03/01/2016] [Indexed: 12/25/2022]
|
45
|
Afizah H, Hui JHP. Mesenchymal stem cell therapy for osteoarthritis. J Clin Orthop Trauma 2016; 7:177-82. [PMID: 27489413 PMCID: PMC4949411 DOI: 10.1016/j.jcot.2016.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 02/08/2023] Open
Abstract
The versatility of mesenchymal stem cells (MSCs) as a treatment modality has landed it another repair target: osteoarthritis, a crippling cartilage disease that frequently afflicts the aged population. Through many studies, this newly discovered method has been shown to significantly alleviate the pain experienced by osteoarthritic patients. Notwithstanding the effectiveness of MSCs in this regard, varying degrees of success rates have also been reported, which is probably attributable to the different approaches adopted in harnessing MSCs' therapeutic value. Accordingly, it is pertinent to understand the contributory factors like MSC type, dosage, size of osteoarthritic lesion, MSC carrier, and mode of infusion, which would be briefly discussed in this review.
Collapse
Affiliation(s)
- Hassan Afizah
- Department of Orthopaedic Surgery, National University Health System, Yong Loo Lin School of Medicine, Singapore,Tissue Engineering Programme, Department of Orthopaedic Surgery, Life Sciences Institute, National University of Singapore, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, National University Health System, Yong Loo Lin School of Medicine, Singapore,Tissue Engineering Programme, Department of Orthopaedic Surgery, Life Sciences Institute, National University of Singapore, Singapore,Corresponding author at: Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block, Level 11, IE Kent Ridge Road, Singapore 119228, Singapore. Tel.: +65 67724321.Department of Orthopaedic Surgery, National University of SingaporeNUHS Tower Block, Level 11, IE Kent Ridge RoadSingapore119228Singapore
| |
Collapse
|
46
|
Zeng Y, Feng S, Liu W, Fu Q, Li Y, Li X, Chen C, Huang C, Ge Z, Du Y. Preconditioning of mesenchymal stromal cells toward nucleus pulposus-like cells by microcryogels-based 3D cell culture and syringe-based pressure loading system. J Biomed Mater Res B Appl Biomater 2015; 105:507-520. [PMID: 26584248 DOI: 10.1002/jbm.b.33509] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/04/2015] [Accepted: 08/14/2015] [Indexed: 01/07/2023]
Abstract
To precondition mesenchymal stromal/stem cells (MSCs) with mechanical stimulation may enhance cell survival and functions following implantation in load bearing environment such as nucleus pulposus (NP) in intervertebral disc (IVD). In this study, preconditioning of MSCs toward NP-like cells was achieved in previously developed poly (ethylene glycol) diacrylate (PEGDA) microcryogels (PMs) within a syringe-based three-dimensional (3D) culture system which provided a facile and cost-effective pressure loading approach. PMs loaded with alginate and MSCs could be incubated in a sealable syringe which could be air-compressed to apply pressure loading through a programmable syringe pump. Expression levels of chondrogenic marker genes SOX9, COL II, and ACAN were significantly upregulated in MSCs when pressure loading of 0.2 MPa or 0.8 MPa was implemented. Expression levels of COL I and COL X were downregulated when pressure loading was applied. In a nude mouse model, MSCs loaded in PMs mechanically stimulated for three days were subcutaneously injected using the same culture syringe. Three weeks postinjection, more proteoglycans (PGs) were deposited and more SOX9 and COL II but less COL I and COL X were stained in 0.2 MPa group. Furthermore, injectable MSCs-loaded PMs were utilized in an ex vivo rabbit IVD organ culture model that demonstrated the leak-proof function and enhanced cell retention of PMs assisted cell delivery to a load bearing environment for potential NP regeneration. This microcryogels-based 3D cell culture and syringe-based pressure loading system represents a novel method for 3D cell culture with mechanical stimulation for better function. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 507-520, 2017.
Collapse
Affiliation(s)
- Yang Zeng
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Siyu Feng
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Qinyouen Fu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yaqian Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Xiaokang Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Chun Chen
- Department of Orthopaedics, Navy General Hospital, Beijing, 100048, China
| | - Chenyu Huang
- Department of Plastic and Reconstructive Surgery, Beijing Tsinghua Changgung Hospital; Medical Center, Tsinghua University, Beijing, 102218, China
| | - Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
47
|
Zeng Y, Zhu L, Han Q, Liu W, Mao X, Li Y, Yu N, Feng S, Fu Q, Wang X, Du Y, Zhao RC. Preformed gelatin microcryogels as injectable cell carriers for enhanced skin wound healing. Acta Biomater 2015; 25:291-303. [PMID: 26234487 DOI: 10.1016/j.actbio.2015.07.042] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 07/18/2015] [Accepted: 07/29/2015] [Indexed: 01/06/2023]
Abstract
Wound dressings of cell-laden bulk hydrogel or scaffold were mainly applied for enhanced cell engraftment in contrast to free cell injection. However, dressing of cells laden in biomaterials on wound surface might not effectively and timely exert functions on deep or chronic wounds where insufficient blood supply exists. Previously, we developed injectable gelatin microcryogels (GMs) which could load cells for enhanced cell delivery and cell therapy. In this study, biological changes of human adipose-derived stem cells (hASCs) laden in GMs were compared in varied aspects with traditional two dimensional (2D) cell culture, such as cell phenotype markers, stemness genes, differentiation, secretion of growth factors, cell apoptosis and cell memory by FACS, QRT-PCR and ELISA, that demonstrated the priming effects of GMs on upregulation of stemness genes and improved secretion of growth factors of hASCs for potential augmented wound healing. In a full-thickness skin wound model in nude mice, multisite injection and dressing of hASCs-laden GMs could significantly accelerate the healing compared to free cell injection. Bioluminescence imaging and protein analysis indicated improved cell retention and secretion of multiple growth factors. Our study suggests that GMs as primed injectable 3D micro-niches represent a new cell delivery methodology for skin wound healing which could not only benefit on the recovery of wound bed but also play direct effects on wound basal layer for healing enhancement. Injectable GMs as facile multisite cell delivery approach potentially provide new minimally-invasive therapeutic strategy for refractory wounds such as diabetic ulcer or radiative skin wound. STATEMENT OF SIGNIFICANCE This work applied a type of elastic micro-scaffold (GMs) to load and prime hMSCs for skin wound healing. Due to the injectability of GMs, the 3D cellular micro-niches could simply realize minimally-invasive and multisite cell delivery approach for accelerating the wound healing process superior to free cell injection. The biological features of MSCs has been thoroughly characterized during 3D culture in GMs (i.e. cell proliferation, characterization of cell surface markers, stemness of MSCs in GMs, differentiation of MSCs in GMs, secretion of MSCs in GMs, induced apoptosis of MSCs in GMs). Multiple methods such as bioluminescent imaging, immunohistochemistry, immunofluorescence, qRT-PCR, ELSA and western blot were used to assess the in vivo results between groups.
Collapse
Affiliation(s)
- Yang Zeng
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Lin Zhu
- Division of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Qin Han
- Center of Excellence in Tissue Engineering, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Xiaojing Mao
- Center of Excellence in Tissue Engineering, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yaqian Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Nanze Yu
- Division of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Siyu Feng
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qinyouen Fu
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiaojun Wang
- Division of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China.
| | - Robert Chunhua Zhao
- Center of Excellence in Tissue Engineering, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Center of Translational Medicine Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
48
|
Qi C, Yan X, Huang C, Melerzanov A, Du Y. Biomaterials as carrier, barrier and reactor for cell-based regenerative medicine. Protein Cell 2015; 6:638-53. [PMID: 26088192 PMCID: PMC4537472 DOI: 10.1007/s13238-015-0179-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/11/2015] [Indexed: 01/24/2023] Open
Abstract
Cell therapy has achieved tremendous success in regenerative medicine in the past several decades. However, challenges such as cell loss, death and immune-rejection after transplantation still persist. Biomaterials have been designed as carriers to deliver cells to desirable region for local tissue regeneration; as barriers to protect transplanted cells from host immune attack; or as reactors to stimulate host cell recruitment, homing and differentiation. With the assistance of biomaterials, improvement in treatment efficiency has been demonstrated in numerous animal models of degenerative diseases compared with routine free cell-based therapy. Emerging clinical applications of biomaterial assisted cell therapies further highlight their great promise in regenerative therapy and even cure for complex diseases, which have been failed to realize by conventional therapeutic approaches.
Collapse
Affiliation(s)
- Chunxiao Qi
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Xiaojun Yan
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Chenyu Huang
- />Department of Plastic and Reconstructive Surgery, Beijing Tsinghua Changgung Hospital; Medical Center, Tsinghua University, Beijing, 102218 China
| | - Alexander Melerzanov
- />Cellular and Molecular Technologies Laboratory, MIPT, Dolgoprudny, 141701 Russia
| | - Yanan Du
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
- />Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310003 China
| |
Collapse
|
49
|
Tseng TC, Tao L, Hsieh FY, Wei Y, Chiu IM, Hsu SH. An Injectable, Self-Healing Hydrogel to Repair the Central Nervous System. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:3518-24. [PMID: 25953204 DOI: 10.1002/adma.201500762] [Citation(s) in RCA: 390] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/04/2015] [Indexed: 05/24/2023]
Abstract
An injectable, self-healing hydrogel (≈1.5 kPa) is developed for healing nerve-system deficits. Neurosphere-like progenitors proliferate in the hydrogel and differentiate into neuron-like cells. In the zebrafish injury model, the central nervous system function is partially rescued by injection of the hydrogel and significantly rescued by injection of the neurosphere-laden hydrogel. The self-healing hydrogel may thus potentially repair the central nervous system.
Collapse
Affiliation(s)
- Ting-Chen Tseng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Fu-Yu Hsieh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan, R.O.C
| | - Shan-hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
50
|
Zeng Y, Chen C, Liu W, Fu Q, Han Z, Li Y, Feng S, Li X, Qi C, Wu J, Wang D, Corbett C, Chan BP, Ruan D, Du Y. Injectable microcryogels reinforced alginate encapsulation of mesenchymal stromal cells for leak-proof delivery and alleviation of canine disc degeneration. Biomaterials 2015; 59:53-65. [PMID: 25956851 DOI: 10.1016/j.biomaterials.2015.04.029] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 04/12/2015] [Accepted: 04/14/2015] [Indexed: 01/07/2023]
Abstract
In situ crosslinked thermo-responsive hydrogel applied for minimally invasive treatment of intervertebral disc degeneration (IVDD) may not prevent extrusion of cell suspension from injection site due to high internal pressure of intervertebral disc (IVD), causing treatment failure or osteophyte formation. In this study, mesenchymal stromal cells (MSCs) were encapsulated in alginate precursor and loaded into previously developed macroporous PGEDA-derived microcryogels (PMs) to form three-dimensional (3D) microscale cellular niches, enabling non-thermo-responsive alginate hydrogel to be injectable. The PMs reinforced alginate hydrogel showed superior elasticity compared to alginate hydrogel alone and could well protect encapsulated cells through injection. Chondrogenic committed MSCs in the injectable microniches expressed higher level of nucleus pulposus (NP) cell markers compared to 2D cultured cells. In an ex vivo organ culture model, injection of MSCs-laden PMs into NP tissue prevented cell leakage, improved cell retention and survival compared to free cell injection. In canine IVDD models, alleviated degeneration was observed in MSCs-laden PMs treated group after six months which was superior to other treated groups. Our results provide in-depth demonstration of injectable alginate hydrogel reinforced by PMs as a leak-proof cell delivery system for augmented regenerative therapy of IVDD in canine models.
Collapse
Affiliation(s)
- Yang Zeng
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Chun Chen
- Department of Orthopaedics, Navy General Hospital, Beijing 100048, China; Department of Orthopedic Surgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Qinyouen Fu
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Zhihua Han
- Department of Orthopaedics, Navy General Hospital, Beijing 100048, China
| | - Yaqian Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Siyu Feng
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiaokang Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Chunxiao Qi
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Jianhong Wu
- Department of Orthopaedics, Navy General Hospital, Beijing 100048, China
| | - Deli Wang
- Department of Orthopaedics, Navy General Hospital, Beijing 100048, China
| | - Christopher Corbett
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Barbara P Chan
- Tissue Engineering Laboratory, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Rd, Hong Kong Special Administrative Region, China
| | - Dike Ruan
- Department of Orthopaedics, Navy General Hospital, Beijing 100048, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China.
| |
Collapse
|