1
|
Zhang X, Zhai H, Zhu X, Geng H, Zhang Y, Cui J, Zhao Y. Polyphenol-Mediated Adhesive and Anti-Inflammatory Double-Network Hydrogels for Repairing Postoperative Intervertebral Disc Defects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53541-53554. [PMID: 39344595 DOI: 10.1021/acsami.4c11901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Hydrogels have garnered tremendous attention for their applications in the repair of intervertebral disk (IVD) degeneration and postoperative IVD defects. However, it is still challenging to develop a hydrogel fulfilling the requirements for high mechanical properties, adhesive capability, biocompatibility, antibacterial properties, and anti-inflammatory performance. Herein, we report a multifunctional double-network (DN) hydrogel composed of physically cross-linked carboxymethyl chitosan (CMCS) and tannic acid (TA) networks as well as chemically cross-linked acrylamide (AM) networks, which integrates the properties of high strength, adhesion, biocompatibility, antimicrobial activity, and anti-inflammation for the repair of postoperative IVD defects. The treatment with CMCS/TA/PAM DN hydrogels can significantly decrease the levels of inflammatory cytokines and degeneration-related factors and upregulated collagen type II alpha 1. In addition, the hydrogels can effectively seal the annulus fibrosus defect, prevent nucleus pulposus degeneration, retain IVD height, and restore the biomechanical properties of the disc to some extent. This polyphenol-mediated DN hydrogel is promising for sealing IVD defects and preventing herniation after lumbar discectomy.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Haoxin Zhai
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xuetao Zhu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Huimin Geng
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Yuanqiang Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Yunpeng Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
2
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
3
|
Liu L, Wang W, Huang L, Xian Y, Ma W, Fan J, Li Y, Liu H, Zheng Z, Wu D. Injectable pathological microenvironment-responsive anti-inflammatory hydrogels for ameliorating intervertebral disc degeneration. Biomaterials 2024; 306:122509. [PMID: 38377847 DOI: 10.1016/j.biomaterials.2024.122509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Chronic local inflammation and resulting cellular dysfunction of nucleus pulposus (NP) cells are important pathogenic factors of intervertebral disc degeneration (IDD). Injectable pathological microenvironment-responsive hydrogels hold significant potential for treating IDD by adapting to dynamic microenvironment of IDD. Herein, we proposed an injectable gelatin-based hydrogel drug delivery system that could respond to the pathological microenvironment of IDD for controlled release of anti-inflammatory drug to promote degenerative NP repair. The hydrogel system was prepared by conjugating phenylboronic acid-modified gelatin methacryloyl (GP) with the naturally extracted anti-inflammatory drug epigallocatechin-3-gallate (EGCG) through dynamic boronic esters. The hydrogel exhibited excellent degradability, injectability, antioxidant properties, anti-inflammatory effects, and biocompatibility. It also displayed responsive-release of EGCG under high reactive oxygen species (ROS) levels and acidic conditions. The hydrogel demonstrated remarkable cytoprotective effects on NP cells in both hyperactive ROS environments and inflammatory cytokine-overexpressed environments in vitro. In vivo studies revealed that the hydrogel injected in situ could effectively ameliorate the intervertebral disc degeneration by maintaining the disc height and NP tissue structure in a rat IDD model. The hydrogel system exhibited excellent biocompatibility and responsive-release of diol-containing drugs in pathological microenvironments, indicating its potential application as a drug delivery platform.
Collapse
Affiliation(s)
- Lei Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wantao Wang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China; Pain Research Center, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Lin Huang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiwen Xian
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wenzheng Ma
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China; Pain Research Center, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jinghao Fan
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yixi Li
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Hongmei Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China; Pain Research Center, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Decheng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
4
|
Qiu R, Cai K, Zhang K, Ying Y, Hu H, Jiang G, Luo K. The current status and development trend of hydrogel application in spinal surgery. J Mater Chem B 2024; 12:1730-1747. [PMID: 38294330 DOI: 10.1039/d3tb02613b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Spinal diseases often result in compromised mobility and diminished quality of life due to the intricate anatomy surrounding the nervous system. Medication and surgical interventions remain the primary treatment methods for spinal conditions. However, currently available medications have limited efficacy in treating spinal surgical diseases and cannot achieve a complete cure. Furthermore, surgical intervention frequently results in inevitable alterations and impairments to the initial anatomical integrity of the spinal structure, accompanied by the consequential loss of certain physiological functionalities. Changes in spine surgery treatment concepts and modalities in the last decade have led to a deepening of minimally invasive treatment, with treatment strategies focusing more on repairing and reconstructing the patient's spine and preserving physiological functions. Therefore, developing novel and more efficient treatment strategies to reduce spinal lesions and iatrogenic injuries is essential. In recent years, significant advancements in biomedical research have led to the discovery that hydrogels possess excellent biocompatibility, biodegradability, and adjustable mechanical properties. The application of hydrogel-based biotechnology in spinal surgery has demonstrated remarkable therapeutic potential. This review presents the therapeutic strategies for spinal diseases based on hydrogel tissue engineering technology.
Collapse
Affiliation(s)
- Rongzhang Qiu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, 315000, China
| | - Kaiwen Cai
- Department of Orthopaedics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315000, China.
| | - Kai Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315000, China.
| | - Yijian Ying
- Health Science Center, Ningbo University, Ningbo, Zhejiang, 315000, China
| | - Hangtian Hu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, 315000, China
| | - Guoqiang Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315000, China.
| | - Kefeng Luo
- Department of Orthopaedics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315000, China.
| |
Collapse
|
5
|
Song H, Guo C, Wu Y, Liu Y, Kong Q, Wang Y. Therapeutic factors and biomaterial-based delivery tools for degenerative intervertebral disc repair. Front Cell Dev Biol 2024; 12:1286222. [PMID: 38374895 PMCID: PMC10875104 DOI: 10.3389/fcell.2024.1286222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is the main cause of low back pain (LBP), which significantly impacts global wellbeing and contributes to global productivity declines. Conventional treatment approaches, encompassing conservative and surgical interventions, merely serve to postpone the advancement of IDD without offering a fundamental reversal. Consequently, there is an urgent demand for an effective approach to prevent the progression of IDD. Recent investigations focusing on the treatment of IDD utilizing diverse bioactive substances integrated within various biomaterials have exhibited promising outcomes. Various bioactive substances, encompassing conventional small molecule drugs, small molecule nucleic acids, and cell therapies, exhibit distinct capacities for repairing IDD. Additionally, various biological material delivery systems, such as nano micelles, microspheres, and hydrogels, possess diverse biological and release characteristics. Consequently, these diverse materials and drugs hold promise for advancing the treatment of IDD. This article aims to provide a concise overview of the IDD process and investigate the research advancements in biomaterials and bioactive substances for IDD treatment, delving into their mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Hao H, Zhao X, Ma H, Lei B, Zhang Y. Oxidative Stress, inflammation and metallomatrix Proteinase-Regulated hydrogels for intervertebral disc regeneration. MATERIALS & DESIGN 2024; 238:112716. [DOI: 10.1016/j.matdes.2024.112716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
7
|
Hu X, Tian X, Yang C, Ling F, Liu H, Zhu X, Pei M, Yang H, Liu T, Xu Y, He F. Melatonin-loaded self-healing hydrogel targets mitochondrial energy metabolism and promotes annulus fibrosus regeneration. Mater Today Bio 2023; 23:100811. [PMID: 37810753 PMCID: PMC10550778 DOI: 10.1016/j.mtbio.2023.100811] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Intervertebral disc (IVD) herniation is a major cause of chronic low back pain and disability. The current nucleus pulposus (NP) discectomy effectively relieves pain symptoms, but the annulus fibrosus (AF) defects are left unrepaired. Tissue engineering approaches show promise in treating AF injury and IVD degeneration; however, the presence of an inflammatory milieu at the injury site hinders the mitochondrial energy metabolism of AF cells, resulting in a lack of AF regeneration. In this study, we fabricated a dynamic self-healing hydrogel loaded with melatonin (an endocrine hormone well-known for its antioxidant and anti-inflammatory properties) and investigate whether melatonin-loaded hydrogel could promote AF defect repair by rescuing the matrix synthesis and energy metabolism of AF cells. The protective effects of melatonin on matrix components (e.g. type I and II collagen and aggrecan) in AF cells were observed in the presence of interleukin (IL)-1β. Additionally, melatonin was found to activate the nuclear factor erythroid 2-related factor signaling pathway, thereby safeguarding the mitochondrial function of AF cells from IL-1β, as evidenced by the increased level of adenosine triphosphate, mitochondrial membrane potential, and respiratory chain factor expression. The incorporation of melatonin into a self-healing hydrogel based on thiolated gelatin and β-cyclodextrin was proposed as a means of promoting AF regeneration. The successful implantation of melatonin-loaded hydrogel has been shown to facilitate in situ regeneration of AF tissue, thereby impeding IVD degeneration by preserving the hydration of nucleus pulposus in a rat box-cut IVD defect model. These findings offer compelling evidence that the development of a melatonin-loaded dynamic self-healing hydrogel can promote the mitochondrial functions of AF cells and represents a promising strategy for IVD regeneration.
Collapse
Affiliation(s)
- Xiayu Hu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Xin Tian
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Chunju Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Feng Ling
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Department of Orthopaedics,the Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Hao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yong Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
- Orthopaedic Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| |
Collapse
|
8
|
Li ZL, Lu Q, Honiball JR, Wan SHT, Yeung KWK, Cheung KMC. Mechanical characterization and design of biomaterials for nucleus pulposus replacement and regeneration. J Biomed Mater Res A 2023; 111:1888-1902. [PMID: 37555381 DOI: 10.1002/jbm.a.37593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/10/2023]
Abstract
Biomaterials for nucleus pulposus (NP) replacement and regeneration have great potential to restore normal biomechanics in degenerated intervertebral discs following nucleotomy. Mechanical characterizations are essential for assessing the efficacy of biomaterial implants for clinical applications. While traditional compression tests are crucial to quantify various modulus values, relaxation behaviors and fatigue resistance, rheological measurements should also be conducted to investigate the viscoelastic properties, injectability, and overall stability upon deformation. To recapitulate the physiological in vivo environment, the use of spinal models is necessary to evaluate the risk of implant extrusion and the restoration of biomechanics under different loading conditions. When designing devices for NP replacement, injectable materials are ideal to fully fill the nucleus cavity and prevent implant migration. In addition to achieving biocompatibility and desirable mechanical characteristics, biomaterial implants should be optimized to avoid implant extrusion or re-herniation post-operatively. This review discusses the most commonly used testing protocols for assessing mechanical properties of biomaterial implants and serves as reference material for enabling researchers to characterize NP implants through a unified approach whereby newly developed biomaterials may be compared and contrasted to existing devices.
Collapse
Affiliation(s)
- Zhuoqi Lucas Li
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Qiuji Lu
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - John Robert Honiball
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Sandra Hiu-Tung Wan
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Kelvin Wai-Kwok Yeung
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Kenneth Man-Chee Cheung
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
9
|
Lu G, Li X, Wang P, Li X, Wang Y, Zhu J, Ronca A, D'Amora U, Liu W, Hui X. Polysaccharide-Based Composite Hydrogel with Hierarchical Microstructure for Enhanced Vascularization and Skull Regeneration. Biomacromolecules 2023; 24:4970-4988. [PMID: 37729544 DOI: 10.1021/acs.biomac.3c00655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Critical-size skull defects caused by trauma, infection, and tumor resection raise great demands for efficient bone substitutes. Herein, a hybrid cross-linked hierarchical microporous hydrogel scaffold (PHCLS) was successfully assembled by a multistep procedure, which involved (i) the preparation of poly(lactic-co-glycolic)/nanohydroxyapatite (PLGA-HAP) porous microspheres, (ii) embedding the spheres in a solution of dopamine-modified hyaluronic acid and collagen I (Col I) and cross-linking via dopamine polyphenols binding to (i) Col I amino groups (via Michael addition) and (ii) PLGA-HAP (via calcium ion chelation). The introduction of PLGA-HAP not only improved the diversity of pore size and pore communication inside the matrix but also greatly enhanced the compressive strength (5.24-fold, 77.5 kPa) and degradation properties to construct a more stable mechanical structure. In particular, the PHCLS (200 mg, nHAP) promoted the proliferation, infiltration, and angiogenic differentiation of bone marrow mesenchymal stem cells in vitro, as well as significant ectopic angiogenesis and mineralization with a storage modulus enhancement of 2.5-fold after 30 days. Meanwhile, the appropriate matrix microenvironment initiated angiogenesis and early osteogenesis by accelerating endogenous stem cell recruitment in situ. Together, the PHCLS allowed substantial skull reconstruction in the rabbit cranial defect model, achieving 85.2% breaking load strength and 84.5% bone volume fractions in comparison to the natural cranium, 12 weeks after implantation. Overall, this study reveals that the hierarchical microporous hydrogel scaffold provides a promising strategy for skull defect treatment.
Collapse
Affiliation(s)
- Gonggong Lu
- Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan 610041, P.R. China
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
| | - Xiang Li
- Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan 610041, P.R. China
| | - Peilei Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
| | - Xing Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
| | - Yuxiang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
| | - Jiayi Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P.R. China
| | - Alfredo Ronca
- National Research Council, Institute of Polymers, Composites and Biomaterials, Naples 80125, Italy
| | - Ugo D'Amora
- National Research Council, Institute of Polymers, Composites and Biomaterials, Naples 80125, Italy
| | - Wenke Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan 610041, P.R. China
| | - Xuhui Hui
- Department of Neurosurgery, West China Hospital, Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
10
|
Pepi S, Paolino M, Saletti M, Venditti J, Talarico L, Andreassi M, Giuliani G, Caselli G, Artusi R, Cappelli A, Leone G, Magnani A, Rovati L. Ferulated Poly(vinyl alcohol) based hydrogels. Heliyon 2023; 9:e22330. [PMID: 38045211 PMCID: PMC10692910 DOI: 10.1016/j.heliyon.2023.e22330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/18/2023] [Accepted: 11/09/2023] [Indexed: 12/05/2023] Open
Abstract
New graft copolymers were prepared by reaction of poly (vinyl alcohol) (PVA) with mono-imidazolide or bis-imidazolide derivatives of ferulic acid (FA) with the formation of ester bonds. The obtained graft copolymers, thanks to the crosslinking capability of FA, formed in water strong gels as verified by rheological analyses. The resulting hydrogels were characterized to evaluate their applicability as wound dressing. In this perspective, their capability to absorb and retain a large amount of fluid without dissolving was verified by swelling kinetics and Moisture Vapour Transmission Rate measurements. Their stability towards mechanical solicitations was assessed by quantifying elasticity, compliance, stress-relaxation, and adhesivity properties. The analyses pointed out that hydrogel PVA-FA2-3 obtained by feruloylation of PVA with bis-imidazole derivative of ferulic acid using an acylation agent/polymer molar ratio 0.03/1 resulted the best candidate for the foreseen application.
Collapse
Affiliation(s)
- Simone Pepi
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Jacopo Venditti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Luigi Talarico
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Marco Andreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | | | - Roberto Artusi
- Rottapharm Biotech, Via Valosa di Sopra 7, 20052, Monza, Italy
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Gemma Leone
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121, Firenze, Italy
| | - Agnese Magnani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università Degli Studi di Siena, Via A. Moro 2, 53100, Siena, Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121, Firenze, Italy
| | - Lucio Rovati
- Rottapharm Biotech, Via Valosa di Sopra 7, 20052, Monza, Italy
| |
Collapse
|
11
|
Bongiovanni Abel S, Busatto CA, Karp F, Estenoz D, Calderón M. Weaving the next generation of (bio)materials: Semi-interpenetrated and interpenetrated polymeric networks for biomedical applications. Adv Colloid Interface Sci 2023; 321:103026. [PMID: 39491440 DOI: 10.1016/j.cis.2023.103026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/01/2023] [Accepted: 10/12/2023] [Indexed: 11/05/2024]
Abstract
Advances in polymer science have led to the development of semi-interpenetrated and interpenetrated networks (SIPN/IPN). The interpenetration procedure allows enhancing several important properties of a polymeric material, including mechanical properties, swelling capability, stimulus-sensitive response, and biological performance, among others. More interestingly, the interpenetration (or semi-interpenetration) can be achieved independent of the material size, that is at the macroscopic, microscopic, or nanometric scale. SIPN/IPN have been used for a wide range of applications, especially in the biomedical field, including tissue engineering, delivery of chemical compounds or biological macromolecules, and multifunctional systems as theragnostic platforms. In the last years, this fascinating field has gained a great interest in the area of polymers for therapeutics; therefore, a comprehensive revision of the topic is timely. In this review, we describe in detail the most relevant synthetic approaches to fabricate polymeric IPN and SIPN, ranging from nanoscale to macroscale. The advantages of typical synthetic methods are analyzed, as well as novel and promising trends in the field of advanced material fabrication. Furthermore, the characterization techniques employed for these materials are summarized from physicochemical, thermal, mechanical, and biological perspectives. The applications of novel (semi-)interpenetrated structures are discussed with a focus on drug delivery, tissue engineering, and regenerative medicine, as well as combinations thereof.
Collapse
Affiliation(s)
- Silvestre Bongiovanni Abel
- Biomedical Polymers Division, INTEMA (National University of Mar del Plata-CONICET), Av. Colón 10850, Mar del Plata 7600, Argentina; POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country, UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain
| | - Carlos A Busatto
- Group of Polymers and Polymerization Reactors, INTEC (National University of Litoral-CONICET), Güemes 3450, Santa Fe 3000, Argentina
| | - Federico Karp
- Group of Polymeric Nanomaterials, INIFTA (National University of La Plata-CONICET), Diagonal 113, La Plata 1900, Argentina
| | - Diana Estenoz
- Group of Polymers and Polymerization Reactors, INTEC (National University of Litoral-CONICET), Güemes 3450, Santa Fe 3000, Argentina
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country, UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain.
| |
Collapse
|
12
|
Seok JM, Kim MJ, Park JH, Kim D, Lee D, Yeo SJ, Lee JH, Lee K, Byun JH, Oh SH, Park SA. A bioactive microparticle-loaded osteogenically enhanced bioprinted scaffold that permits sustained release of BMP-2. Mater Today Bio 2023; 21:100685. [PMID: 37545560 PMCID: PMC10401289 DOI: 10.1016/j.mtbio.2023.100685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/03/2023] [Accepted: 05/29/2023] [Indexed: 08/08/2023] Open
Abstract
Extrusion-based bioprinting technology is widely used for tissue regeneration and reconstruction. However, the method that uses only hydrogel as the bioink base material exhibits limited biofunctional properties and needs improvement to achieve the desired tissue regeneration. In this study, we present a three-dimensionally printed bioactive microparticle-loaded scaffold for use in bone regeneration applications. The unique structure of the microparticles provided sustained release of growth factor for > 4 weeks without the use of toxic or harmful substances. Before and after printing, the optimal particle ratio in the bioink for cell viability demonstrated a survival rate of ≥ 85% over 7 days. Notably, osteogenic differentiation and mineralization-mediated by human periosteum-derived cells in scaffolds with bioactive microparticles-increased over a 2-week interval. Here, we present an alternative bioprinting strategy that uses the sustained release of bioactive microparticles to improve biofunctional properties in a manner that is acceptable for clinical bone regeneration applications.
Collapse
Affiliation(s)
- Ji Min Seok
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min Ji Kim
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jin Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Dahong Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongjin Lee
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Seon Ju Yeo
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Jun Hee Lee
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute for Convergence Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Su A Park
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), Daejeon, 34103, Republic of Korea
| |
Collapse
|
13
|
Chen T, Qian Q, Makvandi P, Zare EN, Chen Q, Chen L, Zhang Z, Zhou H, Zhou W, Wang H, Wang X, Chen Y, Zhou Y, Wu A. Engineered high-strength biohydrogel as a multifunctional platform to deliver nucleic acid for ameliorating intervertebral disc degeneration. Bioact Mater 2023; 25:107-121. [PMID: 37056255 PMCID: PMC10088054 DOI: 10.1016/j.bioactmat.2023.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 02/04/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is a leading cause of low back pain. The strategy of using functional materials to deliver nucleic acids provides a powerful tool for ameliorating IVDD. However, the immunogenicity of nucleic acid vectors and the poor mechanical properties of functional materials greatly limit their effects. Herein, antagomir-204-3p (AM) shows low immunogenicity and effectively inhibits the apoptosis of nucleus pulposus cells. Moreover, a high-strength biohydrogel based on zinc-oxidized sodium alginate-gelatin (ZOG) is designed as a multifunctional nucleic acid delivery platform. ZOG loaded with AM (ZOGA) exhibits great hygroscopicity, antibacterial activity, biocompatibility, and biodegradability. Moreover, ZOGA can be cross-linked with nucleus pulposus tissue to form a high-strength collagen network that improves the mechanical properties of the intervertebral disc (IVD). In addition, ZOGA provides an advantageous microenvironment for genetic expression in which AM can play an efficient role in maintaining the metabolic balance of the extracellular matrix. The results of the radiological and histological analyses demonstrate that ZOGA restores the height of the IVD, retains moisture in the IVD, and maintains the tissue structure. The ZOGA platform shows the sustained release of nucleic acids and has the potential for application to ameliorate IVDD, opening a path for future studies related to IVD.
Collapse
|
14
|
Wang F, Guo K, Nan L, Wang S, Lu J, Wang Q, Ba Z, Huang Y, Wu D. Kartogenin-loaded hydrogel promotes intervertebral disc repair via protecting MSCs against reactive oxygen species microenvironment by Nrf2/TXNIP/NLRP3 axis. Free Radic Biol Med 2023; 204:128-150. [PMID: 37149010 DOI: 10.1016/j.freeradbiomed.2023.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023]
Abstract
Intervertebral disc (IVD) degeneration (IDD) and the consequent low back pain present a major medical challenge. Stem cell-based tissue engineering is promising for the treatment of IDD. However, stem cell-based treatment is severely impaired by the increased generation of reactive oxygen species (ROS) in degenerative disc, which can lead to a high level of cell dysfunction and even death. In this study, a kartogenin (KGN)@PLGA-GelMA/PRP composite hydrogel was designed and used as a carrier of ADSCs-based therapies in disc repair. Injectable composite hydrogel act as a carrier for controlled release of KGN and deliver ADSCs to the degenerative disc. The released KGN can stimulate the differentiation of ADSCs into a nucleus pulposus (NP) -like phenotype and boost antioxidant capacity of ADSCs via activating Nrf2/TXNIP/NLRP3 axis. Furthermore, the composite hydrogel combined with ADSCs attenuated the in vivo degeneration of rat IVDs, maintained IVD tissue integrity and accelerated the synthesis of NP-like extracellular matrix. Therefore, the KGN@PLGA-GelMA/PRP composite hydrogel is a promising strategy for stem cell-based therapies of IDD.
Collapse
Affiliation(s)
- Feng Wang
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Kai Guo
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Liping Nan
- Center for Orthopaedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Shuguang Wang
- Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Jiawei Lu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qiang Wang
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhaoyu Ba
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yufeng Huang
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Desheng Wu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
15
|
Ye J, Liu N, Li Z, Liu L, Zheng M, Wen X, Wang N, Xu Y, Sun B, Zhou Q. Injectable, Hierarchically Degraded Bioactive Scaffold for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:11458-11473. [PMID: 36827205 DOI: 10.1021/acsami.2c18824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Bioactive materials play vital roles in the repair of critical bone defects. However, bone tissue engineering and regenerative medicine are still challenged by the need to repair bone defects evenly and completely. In this study, we functionally simulated the natural creeping substitution process of autologous bone repair by constructing an injectable, hierarchically degradable bioactive scaffold with a composite hydrogel, decalcified bone matrix (DBM) particles, and bone morphogenetic protein 2. This composite scaffold exhibited superior mechanical properties. The scaffold promoted cell proliferation and osteogenic differentiation through multiple signaling pathways. The hierarchical degradation rates of the crosslinked hydrogel and DBM particles accelerated tissue ingrowth and bone formation with a naturally woven bone-like structure in vivo. In the rat calvarial critical defect repair model, the composite scaffold provided even and complete repair of the entire defect area while also integrating the new and host bone effectively. Our results indicate that this injectable, hierarchically degradable bioactive scaffold promotes bone regeneration and provides a promising strategy for evenly and completely repairing the bone defects.
Collapse
Affiliation(s)
- Jixing Ye
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ningyuan Liu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Zongxin Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Liehua Liu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ming Zheng
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xueping Wen
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Nan Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yanqin Xu
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Biemin Sun
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Qiang Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
- Tissue Repair and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| |
Collapse
|
16
|
Ingtipi K, Choudhury BJ, Moholkar VS. Kaolin-embedded cellulose hydrogel with tunable properties as a green fire retardant. Carbohydr Polym 2023; 313:120871. [PMID: 37182962 DOI: 10.1016/j.carbpol.2023.120871] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
This study reports the synthesis of methylene bisacrylamide (MBA) crosslinked cellulose-kaolin (CMK) composite hydrogels. The internal structures of hydrogels were deduced using network parameters, viz. elastic modulus, average molecular weight, mesh size, and effective crosslink. Meanwhile, combustion behavior was investigated using the cone calorimeter test (CCT), limiting oxygen index (LOI) test, vertical flammability test (VFT), and open fire test (OFT). Our results revealed that kaolin addition improves the fire retardancy of hydrogels but reduces their swelling ability. Hydrogel having cellulose to MBA ratio of 1:2 and 2 % w/v kaolin (CM2K2) produced 63 % wt. char residue and the hydrogel-coated cotton fabric exhibited the lowest heat release rate (HRR) of 26.60 kJ/m2 and total heat release (THR) of 0.9 MJ/m2. The LOI of the cotton fabric surged from 20 % to 34.37 % after hydrogel coating. Kinetic analysis using the isoconversional model yielded the highest activation energy (216 kJ/mol) for the CM2K2 hydrogel, corroborating the increased LOI after kaolin addition. VFT and OFT validated the delay in the burning process and the formation of a char layer, which protected the underlying layer of cotton from burning. Overall, cellulose-kaolin hydrogels developed in this study are effective green fire retardant coatings for flammable materials.
Collapse
|
17
|
Lin M, Hu Y, An H, Guo T, Gao Y, Peng K, Zhao M, Zhang X, Zhou H. Silk fibroin-based biomaterials for disc tissue engineering. Biomater Sci 2023; 11:749-776. [PMID: 36537344 DOI: 10.1039/d2bm01343f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Low back pain is the major cause of disability worldwide, and intervertebral disc degeneration (IVDD) is one of the most important causes of low back pain. Currently, there is no method to treat IVDD that can reverse or regenerate intervertebral disc (IVD) tissue, but the recent development of disc tissue engineering (DTE) offers a new means of addressing these disadvantages. Among numerous biomaterials for tissue engineering, silk fibroin (SF) is widely used due to its easy availability and excellent physical/chemical properties. SF is usually used in combination with other materials to construct biological scaffolds or bioactive substance delivery systems, or it can be used alone. The present article first briefly outlines the anatomical and physiological features of IVD, the associated etiology and current treatment modalities of IVDD, and the current status of DTE. Then, it highlights the characteristics of SF biomaterials and their latest research advances in DTE and discusses the prospects and challenges in the application of SF in DTE, with a view to facilitating the clinical process of developing interventions related to IVD-derived low back pain caused by IVDD.
Collapse
Affiliation(s)
- Maoqiang Lin
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China. .,Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| | - Yicun Hu
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China. .,Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| | - Haiying An
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430000, Hubei, China
| | - Taowen Guo
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China. .,Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| | - Yanbing Gao
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China. .,Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| | - Kaichen Peng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China. .,Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| | - Meiling Zhao
- Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| | - Xiaobo Zhang
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China.
| | - Haiyu Zhou
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China. .,Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, Gansu, China
| |
Collapse
|
18
|
He Z, Luo H, Wang Z, Chen D, Feng Q, Cao X. Injectable and tissue adhesive EGCG-laden hyaluronic acid hydrogel depot for treating oxidative stress and inflammation. Carbohydr Polym 2023; 299:120180. [PMID: 36876795 DOI: 10.1016/j.carbpol.2022.120180] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 12/12/2022]
Abstract
Oxidative stress and inflammation are common pathological mechanisms for the progression of tissue degeneration. Epigallocatechin-3-gallate (EGCG) features antioxidant and anti-inflammatory properties, which is a promising drug for the treatment of tissue degeneration. Herein, we utilize the phenylborate ester reaction of EGCG and phenylboronic acid (PBA) to fabricate an injectable and tissue adhesive EGCG-laden hydrogel depot (EGCG HYPOT), which can achieve anti-inflammatory and antioxidative effects via smart delivery of EGCG. Specifically, the phenylborate ester bonds, formed by EGCG and PBA-modified methacrylated hyaluronic acid (HAMA-PBA), endow EGCG HYPOT injectability, shape adaptation and efficient load of EGCG. After photo-crosslinking, EGCG HYPOT exhibits good mechanical properties, tissue adhesion and sustained acid-responsive release of EGCG. EGCG HYPOT can scavenge oxygen and nitrogen free radicals. Meanwhile, EGCG HYPOT can scavenge intracellular reactive oxygen species (ROS) and suppress the expression of pro-inflammatory factors. EGCG HYPOT may provide a new idea for alleviation of inflammatory disturbance.
Collapse
Affiliation(s)
- Zhichao He
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China
| | - Huitong Luo
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, PR China
| | - Zetao Wang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510641, PR China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing 100035, PR China
| | - Qi Feng
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China.
| | - Xiaodong Cao
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510641, PR China.
| |
Collapse
|
19
|
Li Z, Cai F, Tang J, Xu Y, Guo K, Xu Z, Feng Y, Xi K, Gu Y, Chen L. Oxygen metabolism-balanced engineered hydrogel microspheres promote the regeneration of the nucleus pulposus by inhibiting acid-sensitive complexes. Bioact Mater 2022; 24:346-360. [PMID: 36632505 PMCID: PMC9822967 DOI: 10.1016/j.bioactmat.2022.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is commonly caused by imbalanced oxygen metabolism-triggered inflammation. Overcoming the shortcomings of antioxidants in IVDD treatment, including instability and the lack of targeting, remains challenging. Microfluidic and surface modification technologies were combined to graft chitosan nanoparticles encapsulated with strong reductive black phosphorus quantum dots (BPQDs) onto GelMA microspheres via amide bonds to construct oxygen metabolism-balanced engineered hydrogel microspheres (GM@CS-BP), which attenuate extracellular acidosis in nucleus pulposus (NP), block the inflammatory cascade, reduce matrix metalloproteinase expression (MMP), and remodel the extracellular matrix (ECM) in intervertebral discs (IVDs). The GM@CS-BP microspheres reduce H2O2 intensity by 229%. Chemical grafting and electrostatic attraction increase the encapsulation rate of BPQDs by 167% and maintain stable release for 21 days, demonstrating the antioxidant properties and sustained modulation of the BPQDs. After the GM@CS-BP treatment, western blotting revealed decreased acid-sensitive ion channel-3 and inflammatory factors. Histological staining in an 8-week IVDD model confirmed the regeneration of NP. GM@CS-BP microspheres therefore maintain a balance between ECM synthesis and degradation by regulating the positive feedback between imbalanced oxygen metabolism in IVDs and inflammation. This study provides an in-depth interpretation of the mechanisms underlying the antioxidation of BPQDs and a new approach for IVDD treatment.
Collapse
Affiliation(s)
- Ziang Li
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Feng Cai
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Jincheng Tang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yichang Xu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Kaijin Guo
- Department of Orthopedics, the Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, Jiangsu, 221000, PR China
| | - Zonghan Xu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yu Feng
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Kun Xi
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Corresponding author.
| | - Yong Gu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Corresponding author.
| | - Liang Chen
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Corresponding author.
| |
Collapse
|
20
|
Sun D, Liu X, Xu L, Meng Y, Kang H, Li Z. Advances in the Treatment of Partial-Thickness Cartilage Defect. Int J Nanomedicine 2022; 17:6275-6287. [PMID: 36536940 PMCID: PMC9758915 DOI: 10.2147/ijn.s382737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/23/2022] [Indexed: 04/17/2024] Open
Abstract
Partial-thickness cartilage defects (PTCDs) of the articular surface is the most common problem in cartilage degeneration, and also one of the main pathogenesis of osteoarthritis (OA). Due to the lack of a clear diagnosis, the symptoms are often more severe when full-thickness cartilage defect (FTCDs) is present. In contrast to FTCDs and osteochondral defects (OCDs), PTCDs does not injure the subchondral bone, there is no blood supply and bone marrow exudation, and the nearby microenvironment is unsuitable for stem cells adhesion, which completely loses the ability of self-repair. Some clinical studies have shown that partial-thickness cartilage defects is as harmful as full-thickness cartilage defects. Due to the poor effect of conservative treatment, the destructive surgical treatment is not suitable for the treatment of partial-thickness cartilage defects, and the current tissue engineering strategies are not effective, so it is urgent to develop novel strategies or treatment methods to repair PTCDs. In recent years, with the interdisciplinary development of bioscience, mechanics, material science and engineering, many discoveries have been made in the repair of PTCDs. This article reviews the current status and research progress in the treatment of PTCDs from the aspects of diagnosis and modeling of PTCDs, drug therapy, tissue transplantation repair technology and tissue engineering ("bottom-up").
Collapse
Affiliation(s)
- Daming Sun
- Wuhan Sports University, Wuhan, People’s Republic of China
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Xiangzhong Liu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Liangliang Xu
- Wuhan Sports University, Wuhan, People’s Republic of China
| | - Yi Meng
- Wuhan Sports University, Wuhan, People’s Republic of China
| | - Haifei Kang
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, People’s Republic of China
| | - Zhanghua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
21
|
Demott CJ, Grunlan MA. Emerging polymeric material strategies for cartilage repair. J Mater Chem B 2022; 10:9578-9589. [PMID: 36373438 DOI: 10.1039/d2tb02005j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cartilage is found throughout the body, serving an array of essential functions. Owing to the limited healing capacity of cartilage, damage or degeneration is often permanent and so requires clinical intervention. Established surgical techniques generally rely on biological grafting. However, recent advances in polymeric materials provide an encouraging alternative to overcome limits of auto- and allografts. For regenerative engineering of cartilage, a polymeric scaffold ideally supports and instructs tissue regeneration while also providing mechanical integrity. Scaffolds direct regeneration via chemical and mechanical cues, as well as delivery and support of exogenous cells and bioactive factors. Advanced polymeric scaffolds aim to direct regeneration locally, replicating the heterogeneities of native tissues. Alternatively, new cartilage-mimetic hydrogels have potential to serve as synthetic cartilage replacements. Prepared as multi-network or composite hydrogels, the most promising candidates have simultaneously realized the hydration, mechanical, and tribological properties of native cartilage. Collectively, the recent rise in polymers for cartilage regeneration and replacement proposes a changing paradigm, with a new generation of materials paving the way for improved clinical outcomes.
Collapse
Affiliation(s)
- Connor J Demott
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3003, USA
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3003, USA.,Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77843-3003, USA.,Department of Chemistry, Texas A&M University, College Station, TX 77843-3003, USA.
| |
Collapse
|
22
|
Yang L, Yu C, Fan X, Zeng T, Yang W, Xia J, Wang J, Yao L, Hu C, Jin Y, Zhu Y, Chen J, Hu Z. Dual-dynamic-bond cross-linked injectable hydrogel of multifunction for intervertebral disc degeneration therapy. J Nanobiotechnology 2022; 20:433. [PMID: 36182921 PMCID: PMC9526989 DOI: 10.1186/s12951-022-01633-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/06/2022] [Indexed: 11/12/2022] Open
Abstract
Developing smart hydrogels with integrated and suitable properties to treat intervertebral disc degeneration (IVDD) by minimally invasive injection is of high desire in clinical application and still an ongoing challenge. In this work, an extraordinary injectable hydrogel PBNPs@OBG (Prussian blue nanoparticles@oxidized hyaluronic acid/borax/gelatin) with promising antibacterial, antioxidation, rapid gelation, and self-healing characteristics was designed via dual-dynamic-bond cross-linking among the oxidized hyaluronic acid (OHA), borax, and gelatin. The mechanical performance of the hydrogel was studied by dynamic mechanical analysis. Meanwhile, the swelling ratio and degradation level of the hydrogel was explored. Benefiting from its remarkable mechanical properties, sufficient tissue adhesiveness, and ideal shape-adaptability, the injectable PBNPs containing hydrogel was explored for IVDD therapy. Astoundingly, the as-fabricated hydrogel was able to alleviate H2O2-induced excessive ROS against oxidative stress trauma of nucleus pulposus, which was further revealed by theoretical calculations. Rat IVDD model was next established to estimate therapeutic effect of this PBNPs@OBG hydrogel for IVDD treatment in vivo. On the whole, combination of the smart multifunctional hydrogel and nanotechnology-mediated antioxidant therapy can serve as a fire-new general type of therapeutic strategy for IVDD and other oxidative stress-related diseases.
Collapse
Affiliation(s)
- Linjun Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, People's Republic of China
| | - Tianni Zeng
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Hangzhou, 310007, People's Republic of China
| | - Wentao Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Jiechao Xia
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Jianle Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Litao Yao
- Department of Dentistry, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Chuan Hu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Yang Jin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Yutao Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Jiaxin Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China
| | - Zhijun Hu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Key Laboratory of Musculoskeletal System Degeneration, Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 East Qing Chun Road, Hangzhou, 310002, People's Republic of China.
| |
Collapse
|
23
|
Hu J, Li C, Jin S, Ye Y, Fang Y, Xu P, Zhang C. Salvianolic acid B combined with bone marrow mesenchymal stem cells piggybacked on HAMA hydrogel re-transplantation improves intervertebral disc degeneration. Front Bioeng Biotechnol 2022; 10:950625. [PMID: 36237221 PMCID: PMC9552300 DOI: 10.3389/fbioe.2022.950625] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cell-based tissue engineering approaches have emerged as a realistic alternative for regenerative disc tissue repair. The multidirectional differentiation potential of bone marrow mesenchymal stem cells (BMSCs) to treat disc degeneration intervertebral disc degeneration has also become a viable option. We used 1% HAMA hydrogel as a carrier and co-encapsulated BMSCs and Salvianolic acid B (SalB) into the hydrogel to reduce the apoptosis of the transplanted cells. The protective effect of SalB on BMSCs was first verified in vitro using the CCK8 method, flow cytometry, and Western-Blotting, and the physical properties and biocompatibility of HAMA hydrogels were verified in vitro. The rat model was then established using the pinprick method and taken at 4 and 8 W, to examine the extent of disc degeneration by histology and immunohistochemistry, respectively. It was found that SalB could effectively reduce the apoptosis of BMSCs in vitro by activating the JAK2-STAT3 pathway. 1% HAMA hydrogels had larger pore size and better water retention, and the percentage of cell survival within the hydrogels was significantly higher after the addition of SalB to the HAMA hydrogels. In the in vivo setting, the HAMA + SalB + BMSCs group had a more pronounced delaying effect on the progression of disc degeneration compared to the other treatment groups. The method used in this study to encapsulate protective drugs with stem cells in a hydrogel for injection into the lesion has potential research value in the field of regenerative medicine.
Collapse
Affiliation(s)
- Jie Hu
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Key Laboratory of Tissue Transplantation in Anhui Province, Bengbu Medical College, Bengbu, Anhui, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Cai Li
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Key Laboratory of Tissue Transplantation in Anhui Province, Bengbu Medical College, Bengbu, Anhui, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Shichang Jin
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Yuchen Ye
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Yuekun Fang
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Key Laboratory of Tissue Transplantation in Anhui Province, Bengbu Medical College, Bengbu, Anhui, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Panpan Xu
- Bengbu Medical College, Bengbu, Anhui, China
| | - Changchun Zhang
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Key Laboratory of Tissue Transplantation in Anhui Province, Bengbu Medical College, Bengbu, Anhui, China
- Bengbu Medical College, Bengbu, Anhui, China
- *Correspondence: Changchun Zhang,
| |
Collapse
|
24
|
Zhao X, Zhao W, Zhang Y, Zhang X, Ma Z, Wang R, Wei Q, Ma S, Zhou F. Recent progress of bioinspired cartilage hydrogel lubrication materials. BIOSURFACE AND BIOTRIBOLOGY 2022. [DOI: 10.1049/bsb2.12047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Xiaoduo Zhao
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
- Yantai Zhongke Research Institute of Advanced Materials and Green Chemical Engineering Yantai China
| | - Weiyi Zhao
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
| | - Yunlei Zhang
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
| | - Xiaoqing Zhang
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
| | - Zhengfeng Ma
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
- Baiyin Zhongke Innovation Research Institute of Green Materials Baiyin China
| | - Rui Wang
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
| | - Qiangbing Wei
- College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou China
| | - Shuanhong Ma
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
- Yantai Zhongke Research Institute of Advanced Materials and Green Chemical Engineering Yantai China
| | - Feng Zhou
- State Key Laboratory of Solid Lubrication Lanzhou Institute of Chemical Physics Chinese Academy of Sciences Lanzhou China
| |
Collapse
|
25
|
Saletti M, Paolino M, Ballerini L, Giuliani G, Leone G, Lamponi S, Andreassi M, Bonechi C, Donati A, Piovani D, Schieroni AG, Magnani A, Cappelli A. Click-Chemistry Cross-Linking of Hyaluronan Graft Copolymers. Pharmaceutics 2022; 14:pharmaceutics14051041. [PMID: 35631626 PMCID: PMC9146110 DOI: 10.3390/pharmaceutics14051041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 01/27/2023] Open
Abstract
An easy and viable crosslinking procedure by click-chemistry (click-crosslinking) of hyaluronic acid (HA) was developed. In particular, the clickable propargyl groups of hyaluronane-based HA-FA-Pg graft copolymers showing low and medium molecular weight values were exploited in crosslinking by click-chemistry by using a hexa(ethylene glycol) spacer. The resulting HA-FA-HEG-CL materials showed an apparent lack of in vitro cytotoxic effects, tuneable water affinity, and rheological properties according to the crosslinking degree that suggests their applicability in different biomedical fields.
Collapse
Affiliation(s)
- Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
- Correspondence: (M.P.); (A.C.); Tel.: +39-0577-234320 (A.C.)
| | - Lavinia Ballerini
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Gemma Leone
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Stefania Lamponi
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Marco Andreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Claudia Bonechi
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Alessandro Donati
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Daniele Piovani
- Istituto di Scienze e Tecnologie Chimiche “G. Natta”-SCITEC (CNR), Via A. Corti 12, 20133 Milano, Italy; (D.P.); (A.G.S.)
| | - Alberto Giacometti Schieroni
- Istituto di Scienze e Tecnologie Chimiche “G. Natta”-SCITEC (CNR), Via A. Corti 12, 20133 Milano, Italy; (D.P.); (A.G.S.)
| | - Agnese Magnani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018–2022), Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (L.B.); (G.G.); (G.L.); (S.L.); (M.A.); (C.B.); (A.D.); (A.M.)
- Correspondence: (M.P.); (A.C.); Tel.: +39-0577-234320 (A.C.)
| |
Collapse
|
26
|
Koetsier E, van Kuijk SMJ, Maino P, Dukanac J, Scascighini L, Cianfoni A, Scarone P, Kuhlen DE, Hollman MW, Kallewaard JW. Efficacy of the Gelstix nucleus augmentation device for the treatment of chronic discogenic low back pain: protocol for a randomised, sham-controlled, double-blind, multicentre trial. BMJ Open 2022; 12:e053772. [PMID: 35354635 PMCID: PMC8968527 DOI: 10.1136/bmjopen-2021-053772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Discogenic pain is the cause of pain in 26%-40% of patients with for low back pain. Consensus about treatment of chronic discogenic low back pain is lacking and most treatment alternatives are supported by limited evidence. The percutaneous implantation of hydrogels into the nucleus pulposus represents a promising regenerative intradiscal therapy. The hydrogel 'GelStix' is composed primarily of hydrolyzed polyacrylonitrile and acts as a reservoir of hydration, producing increased pressure and improved pH balance, potentially leading to disc preservation. We hypothesise that treatment with GelStix will lead to greater reduction in pain intensity at 6 months post-treatment compared with patients receiving sham treatment. METHODS AND ANALYSIS This is a parallel group, randomised sham-controlled double-blind, multicentre trial to assess whether the GelStix device is superior to sham in reducing pain intensity in patients with chronic discogenic low back pain. The study will be conducted in two regional hospitals in Europe. Seventy-two participants will be randomised in a 1:1 ratio. The primary outcome will be the change in pain intensity between preoperative baseline and at 6 months postintervention. Secondary outcomes were disability, quality of life, the patient's global impression of change scale, the use of pain medication and the disc degeneration process assessed by means of MRI. For change in pain intensity, disability, health-related quality of life and disc height, mean values will be compared between groups using linear regression analysis, adjusted for treatment centre. ETHICS AND DISSEMINATION Ethics approval was obtained from the Ethics Committee of the Canton Ticino, Switzerland (CE2982) and by the Medical Ethical Committee Arnhem-Nijmegen, the Netherlands (2016-2944). All patients that agree to participate will be asked to sign an informed consent form. Results will be disseminated through international publications in peer-reviewed journals, in addition to international conference presentations. TRIAL REGISTRATION NUMBER NCT02763956. PROTOCOL VERSION 7.1, 18 November 2020.
Collapse
Affiliation(s)
- Eva Koetsier
- Pain Management Center, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Sander M J van Kuijk
- Clinical Epidemiology and Medical Technology Assessment, Maastricht Universitair Medisch Centrum+, Maastricht, The Netherlands
| | - Paolo Maino
- Pain Management Center, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Jasmina Dukanac
- Pain Management Center, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
| | - Luca Scascighini
- Department of Health Sciences, University of Applied Sciences and Arts of Southern Switzerland, Manno, Switzerland
| | - Alessandro Cianfoni
- Service of Diagnostic and Interventional Neuroradiology, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
- Department of Neuroradiology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Pietro Scarone
- Clinic of Neurosurgery, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
| | - Dominique E Kuhlen
- Clinic of Neurosurgery, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
| | - Markus W Hollman
- Department of Anesthesiology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | | |
Collapse
|
27
|
Kamatani T, Hagizawa H, Yarimitsu S, Morioka M, Koyamatsu S, Sugimoto M, Kodama J, Yamane J, Ishiguro H, Shichino S, Abe K, Fujibuchi W, Fujie H, Kaito T, Tsumaki N. Human iPS cell-derived cartilaginous tissue spatially and functionally replaces nucleus pulposus. Biomaterials 2022; 284:121491. [PMID: 35395453 DOI: 10.1016/j.biomaterials.2022.121491] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 12/12/2022]
Abstract
The loss of nucleus pulposus (NP) precedes the intervertebral disk (IVD) degeneration that causes back pain. Here, we demonstrate that the implantation of human iPS cell-derived cartilaginous tissue (hiPS-Cart) restores this loss by replacing lost NP spatially and functionally. NP cells consist of notochordal NP cells and chondrocyte-like NP cells. Single cell RNA sequencing (scRNA-seq) analysis revealed that cells in hiPS-Cart corresponded to chondrocyte-like NP cells but not to notochordal NP cells. The implantation of hiPS-Cart into a nuclectomized space of IVD in nude rats prevented the degeneration of the IVD and preserved its mechanical properties. hiPS-Cart survived and occupied the nuclectomized space for at least six months after implantation, indicating spatial and functional replacement of lost NP by hiPS-Cart. Further scRNA-seq analysis revealed that hiPS-Cart cells changed their profile after implantation, differentiating into two lineages that are metabolically distinct from each other. However, post-implanted hiPS-Cart cells corresponded to chondrocyte-like NP cells only and did not develop into notochordal NP cells, suggesting that chondrocyte-like NP cells are nearly sufficient for NP function. The data collectively indicate that hiPS-Cart is a candidate implant for regenerating NP spatially and functionally and preventing IVD degeneration.
Collapse
Affiliation(s)
- Takashi Kamatani
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroki Hagizawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Seido Yarimitsu
- Department of Mechanical Systems Engineering, Faculty of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Miho Morioka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Saeko Koyamatsu
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michihiko Sugimoto
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Joe Kodama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Junko Yamane
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroyuki Ishiguro
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Building 17 Second Floor, 2641, Yamasaki, Noda, Chiba, 278-0042, Japan
| | - Kuniya Abe
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Wataru Fujibuchi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiromichi Fujie
- Department of Mechanical Systems Engineering, Faculty of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Tsumaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
28
|
Hickman TT, Rathan-Kumar S, Peck SH. Development, Pathogenesis, and Regeneration of the Intervertebral Disc: Current and Future Insights Spanning Traditional to Omics Methods. Front Cell Dev Biol 2022; 10:841831. [PMID: 35359439 PMCID: PMC8963184 DOI: 10.3389/fcell.2022.841831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
The intervertebral disc (IVD) is the fibrocartilaginous joint located between each vertebral body that confers flexibility and weight bearing capabilities to the spine. The IVD plays an important role in absorbing shock and stress applied to the spine, which helps to protect not only the vertebral bones, but also the brain and the rest of the central nervous system. Degeneration of the IVD is correlated with back pain, which can be debilitating and severely affects quality of life. Indeed, back pain results in substantial socioeconomic losses and healthcare costs globally each year, with about 85% of the world population experiencing back pain at some point in their lifetimes. Currently, therapeutic strategies for treating IVD degeneration are limited, and as such, there is great interest in advancing treatments for back pain. Ideally, treatments for back pain would restore native structure and thereby function to the degenerated IVD. However, the complex developmental origin and tissue composition of the IVD along with the avascular nature of the mature disc makes regeneration of the IVD a uniquely challenging task. Investigators across the field of IVD research have been working to elucidate the mechanisms behind the formation of this multifaceted structure, which may identify new therapeutic targets and inform development of novel regenerative strategies. This review summarizes current knowledge base on IVD development, degeneration, and regenerative strategies taken from traditional genetic approaches and omics studies and discusses the future landscape of investigations in IVD research and advancement of clinical therapies.
Collapse
Affiliation(s)
- Tara T. Hickman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sudiksha Rathan-Kumar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sun H. Peck
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
- *Correspondence: Sun H. Peck,
| |
Collapse
|
29
|
Liu Z, Wei H, Liu Y, Li W, Li S, Wang G, Guo T. Fabrication and characterization of interpenetrating network hydrogels based on sequential amine‐anhydride reaction and photopolymerization in water. POLYM ENG SCI 2022. [DOI: 10.1002/pen.25896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Zijun Liu
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| | - Hongliang Wei
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| | - Yuhua Liu
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| | - Weikun Li
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| | - Songmao Li
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| | - Gang Wang
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| | - Tao Guo
- School of Chemistry and Chemical Engineering Henan University of Technology Zhengzhou PR China
| |
Collapse
|
30
|
Ling Z, Liu Y, Wang Z, Zhang Z, Chen B, Yang J, Zeng B, Gao Y, Jiang C, Huang Y, Zou X, Wang X, Wei F. Single-Cell RNA-Seq Analysis Reveals Macrophage Involved in the Progression of Human Intervertebral Disc Degeneration. Front Cell Dev Biol 2022; 9:833420. [PMID: 35295968 PMCID: PMC8918513 DOI: 10.3389/fcell.2021.833420] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Intervertebral disc degeneration (IDD) has been considered as the primary pathological mechanism that underlies low back pain. Understanding the molecular mechanisms underlying human IDD is imperative for making strategies to treat IDD-related diseases. Herein, we report the molecular programs, lineage progression patterns, and paths of cellular communications during the progression of IDD using single-cell RNA sequencing (scRNA-seq) on nucleus pulposus (NP) cells from patients with different grades of IDD undergoing discectomy. New subtypes of cells and cell-type-specific gene signatures of the metabolic homeostatic NP cells (Met NPC), adhesive NP cells (Adh NPC), inflammatory response NP cells (IR NPC), endoplasmic reticulum stress NP cells (ERS NPC), fibrocartilaginous NP cells (Fc NPC), and CD70 and CD82+ progenitor NP cells (Pro NPC) were identified. In the late stage of IDD, the IR NPC and Fc NPC account for a large proportion of NPC. Importantly, immune cells including macrophages, T cells, myeloid progenitors, and neutrophils were also identified, and further analysis showed that significant intercellular interaction between macrophages and Pro NPC occurred via MIF (macrophage migration inhibitory factor) and NF-kB signaling pathways during the progression of IDD. In addition, dynamic polarization of macrophage M1 and M2 cell subtypes was found in the progression of IDD, and gene set functional enrichment analysis suggested a significant role of the macrophage polarization in regulating cell metabolism, especially the Pro NPC. Finally, we found that the NP cells in the late degenerative stage were mainly composed of the cell types related to inflammatory and endoplasmic reticulum (ER) response, and fibrocartilaginous activity. Our results provided new insights into the identification of NP cell populations at single-cell resolution and at the relatively whole-transcriptome scale, accompanied by cellular communications between immune cells and NP cells, and discriminative markers in relation to specific cell subsets. These new findings present clues for effective and functional manipulation of human IDD-related bioremediation and healthcare.
Collapse
Affiliation(s)
- Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Liu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bolin Chen
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiaming Yang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Baozhu Zeng
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yu Gao
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chang Jiang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yulin Huang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiuhui Wang
- Department of Orthopaedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Fuxin Wei, ; Xiuhui Wang,
| | - Fuxin Wei
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Fuxin Wei, ; Xiuhui Wang,
| |
Collapse
|
31
|
Guo W, Douma L, Hu MH, Eglin D, Alini M, Šećerović A, Grad S, Peng X, Zou X, D'Este M, Peroglio M. Hyaluronic acid-based interpenetrating network hydrogel as a cell carrier for nucleus pulposus repair. Carbohydr Polym 2022; 277:118828. [PMID: 34893245 DOI: 10.1016/j.carbpol.2021.118828] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/08/2021] [Accepted: 10/27/2021] [Indexed: 01/19/2023]
Abstract
Hyaluronic acid (HA) is a key component of the intervertebral disc (IVD) that is widely investigated as an IVD biomaterial. One persisting challenge is introducing materials capable of supporting cell encapsulation and function, yet with sufficient mechanical stability. In this study, a hybrid interpenetrating polymer network (IPN) was produced as a non-covalent hydrogel, based on a covalently cross-linked HA (HA-BDDE) and HA-poly(N-isopropylacrylamide) (HA-pNIPAM). The hybrid IPN was investigated for its physicochemical properties, with histology and gene expression analysis to determine matrix deposition in vitro and in an ex vivo model. The IPN hydrogel displayed cohesiveness for at least one week and rheological properties resembling native nucleus pulposus (NP) tissue. When implanted in an ex vivo IVD organ culture model, the IPN supported cell viability, phenotype expression of encapsulated NP cells and IVD matrix production over four weeks under physiological loading. Overall, our results indicate the therapeutic potential of this HA-based IPN hydrogel for IVD regeneration.
Collapse
Affiliation(s)
- Wei Guo
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland; Department of Spinal Surgery, Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Luzia Douma
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Ming Hsien Hu
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Amra Šećerović
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Sibylle Grad
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Xinsheng Peng
- Department of Spinal Surgery, Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Xuenong Zou
- Department of Spinal Surgery, Orthopaedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland.
| | - Marianna Peroglio
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| |
Collapse
|
32
|
Culbert MP, Warren JP, Dixon AR, Fermor HL, Beales PA, Wilcox RK. Evaluation of injectable nucleus augmentation materials for the treatment of intervertebral disc degeneration. Biomater Sci 2021; 10:874-891. [PMID: 34951410 DOI: 10.1039/d1bm01589c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Back pain affects a person's health and mobility as well as being associated with large health and social costs. Lower back pain is frequently caused by degeneration of the intervertebral disc. Current operative and non-operative treatments are often ineffective and expensive. Nucleus augmentation is designed to be a minimally invasive method of restoring the disc to its native healthy state by restoring the disc height, and mechanical and/or biological properties. The majority of the candidate materials for nucleus augmentation are injectable hydrogels. In this review, we examine the materials that are currently under investigation for nucleus augmentation, and compare their ability to meet the design requirements for this application. Specifically, the delivery of the material into the disc, the mechanical properties of the material and the biological compatibility are examined. Recommendations for future testing are also made.
Collapse
Affiliation(s)
- Matthew P Culbert
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - James P Warren
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - Andrew R Dixon
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - Hazel L Fermor
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - Paul A Beales
- School of Chemistry, Astbury Centre for Structural Molecular Biology and Bragg Centre for Materials Research, University of Leeds, UK, LS2 9JT
| | - Ruth K Wilcox
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| |
Collapse
|
33
|
Insight into the pharmacological effects of andrographolide in musculoskeletal disorders. Biomed Pharmacother 2021; 146:112583. [PMID: 34954644 DOI: 10.1016/j.biopha.2021.112583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/23/2022] Open
Abstract
Andrographis paniculata (A. paniculata) is a traditional herbal medicine that has been widely used in Asian countries for hundreds of years. Andrographolide (AG) is a diterpene lactone extracted from A. paniculata. Owing to the in-depth study of pharmacological mechanisms, the therapeutic potential of AG, including its anti-inflammatory, anti-tumor, and immunoregulatory attributes, has attracted the attention of many researchers. Studies testing the therapeutic effects of AG have demonstrated desirable results in the treatment of a variety of clinical diseases. With high safety and various biological functions, AG might be a promising candidate for the treatment of musculoskeletal disorders. Here, we review all available literatures to summarize the pharmacological effects of AG and facilitate further researches on musculoskeletal diseases.
Collapse
|
34
|
Fu H, Yu C, Li X, Bao H, Zhang B, Chen Z, Zhang Z. Facile engineering of ECM-mimetic injectable dual crosslinking hydrogels with excellent mechanical resilience, tissue adhesion, and biocompatibility. J Mater Chem B 2021; 9:10003-10014. [PMID: 34874044 DOI: 10.1039/d1tb01914g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Injectable hydrogels have aroused ever-increasing interest for their cell/biomaterial delivery ability through minimally invasive procedures. Nevertheless, it is still a challenge to simply fabricate natural biopolymer-based injectable hydrogels possessing satisfactory mechanical properties, bioadhesion, and cell delivery ability. Herein, we describe a facile dual crosslinking (DC) strategy for preparing extracellular matrix (ECM) mimetic hydrogels with desirable comprehensive performance. The chondroitin sulfate (CS)- and gelatin (Gel)-based single crosslinked (SC) hydrogels were first developed via reversible borate ester bonds, and further strengthened through the Michael-addition crosslinking reaction or visible-light initiated photopolymerization with thiol-containing polyethylene glycol (PEG) crosslinkers. The dynamic SC hydrogels showed good injectability, pH-sensitive gel-sol transformation, and self-adhesion ability to various biological tissues such as skin, liver, and intervertebral disc. The mechanically tough DC hydrogels displayed tunable stiffness, and resilience to compression load (up to 90% strain) owing to the effective energy dissipation mechanism. The formed DC hydrogels after subcutaneous injection well integrated with surrounding tissues and exhibited fast self-recovery properties. Moreover, the photoencapsulation of human mesenchymal stem cells (hMSCs) within the developed DC hydrogels was achieved and has been proved to be biocompatible, highlighting the great potential of the photopolymerized DC hydrogels in cell delivery and three-dimensional (3D) cell culture. This biomimetic, mechanically resilient, adhesive, and cytocompatible injectable DC hydrogel could serve as a promising candidate for tissue engineering.
Collapse
Affiliation(s)
- Han Fu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China. .,University of Chinese Academy of Sciences, 19(A) Yuquan Road, Beijing 100049, China
| | - Chenggong Yu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Xiaodi Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Hongying Bao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Bo Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Zhongjin Chen
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Zhijun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nanobionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
35
|
Malli SE, Kumbhkarn P, Dewle A, Srivastava A. Evaluation of Tissue Engineering Approaches for Intervertebral Disc Regeneration in Relevant Animal Models. ACS APPLIED BIO MATERIALS 2021; 4:7721-7737. [PMID: 35006757 DOI: 10.1021/acsabm.1c00500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Translation of tissue engineering strategies for the regeneration of intervertebral disc (IVD) requires a strong understanding of pathophysiology through the relevant animal model. There is no relevant animal model due to differences in disc anatomy, cellular composition, extracellular matrix components, disc physiology, and mechanical strength from humans. However, available animal models if used correctly could provide clinically relevant information for the translation into humans. In this review, we have investigated different types of strategies for the development of clinically relevant animal models to study biomaterials, cells, biomolecular or their combination in developing tissue engineering-based treatment strategies. Tissue engineering strategies that utilize various animal models for IVD regeneration are summarized and outcomes have been discussed. The understanding of animal models for the validation of regenerative approaches is employed to understand and treat the pathophysiology of degenerative disc disease (DDD) before proceeding for human trials. These animal models play an important role in building a therapeutic regime for IVD tissue regeneration, which can serve as a platform for clinical applications.
Collapse
Affiliation(s)
- Sweety Evangeli Malli
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-Ahmedabad), Gandhinagar, Gujarat 382355, India
| | - Pranav Kumbhkarn
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-Ahmedabad), Gandhinagar, Gujarat 382355, India
| | - Ankush Dewle
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-Ahmedabad), Gandhinagar, Gujarat 382355, India
| | - Akshay Srivastava
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-Ahmedabad), Gandhinagar, Gujarat 382355, India
| |
Collapse
|
36
|
Li C, Bai Q, Lai Y, Tian J, Li J, Sun X, Zhao Y. Advances and Prospects in Biomaterials for Intervertebral Disk Regeneration. Front Bioeng Biotechnol 2021; 9:766087. [PMID: 34746112 PMCID: PMC8569141 DOI: 10.3389/fbioe.2021.766087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Low-back and neck-shoulder pains caused by intervertebral disk degeneration are highly prevalent among middle-aged and elderly people globally. The main therapy method for intervertebral disk degeneration is surgical intervention, including interbody fusion, disk replacement, and diskectomy. However, the stress changes caused by traditional fusion surgery are prone to degeneration of adjacent segments, while non-fusion surgery has problems, such as ossification of artificial intervertebral disks. To overcome these drawbacks, biomaterials that could endogenously regenerate the intervertebral disk and restore the biomechanical function of the intervertebral disk is imperative. Intervertebral disk is a fibrocartilaginous tissue, primarily comprising nucleus pulposus and annulus fibrosus. Nucleus pulposus (NP) contains high water and proteoglycan, and its main function is absorbing compressive forces and dispersing loads from physical activities to other body parts. Annulus fibrosus (AF) is a multilamellar structure that encloses the NP, comprises water and collagen, and supports compressive and shear stress during complex motion. Therefore, different biomaterials and tissue engineering strategies are required for the functional recovery of NP and AF based on their structures and function. Recently, great progress has been achieved on biomaterials for NP and AF made of functional polymers, such as chitosan, collagen, polylactic acid, and polycaprolactone. However, scaffolds regenerating intervertebral disk remain unexplored. Hence, several tissue engineering strategies based on cell transplantation and growth factors have been extensively researched. In this review, we summarized the functional polymers and tissue engineering strategies of NP and AF to endogenously regenerate degenerative intervertebral disk. The perspective and challenges of tissue engineering strategies using functional polymers, cell transplantation, and growth factor for generating degenerative intervertebral disks were also discussed.
Collapse
Affiliation(s)
- Chunxu Li
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qiushi Bai
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuxiao Lai
- Centre for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jingjing Tian
- Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiahao Li
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Yu Zhao
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Wei C, Song J, Tan H. A paintable ophthalmic adhesive with customizable properties based on symmetrical/asymmetrical cross-linking. Biomater Sci 2021; 9:7522-7533. [PMID: 34643623 DOI: 10.1039/d1bm01197a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In situ and efficient incision sealing for ophthalmic surgery remains unresolved. Current commercially available gel adhesives often suffer from unsuitable gelation time, difficulty in micro-delivery, and mismatched degradation period, leading to difficulties for application in ocular tissue areas. Herein, a novel hydrogel adhesive was developed based on the simultaneous crosslinking of poly(lysine) (PLL) and lysine (Lys) with an end-modified active ester multi-arm polyethylene glycol (PEG) via the amidation reaction, where the residual terminal active ester of PEG can also bond to amino groups on tissue to provide strong adhesion. Due to the different molecular structures around their amino groups, PLL and Lys can crosslink with 4-arm-PEG-NHS (active ester) respectively, to form symmetrical and asymmetrical crosslinking networks, which exhibit various mechanical properties. Therefore, just by adjusting PLL/Lys ratios, the PEG-PLL-Lys hydrogel can easily possess a suitable gelation time, appropriate mechanical properties and matched degradation rate. As a result, a paintable, readily accessible and biocompatible ophthalmic tissue adhesive (sealant) is prepared for sealing ocular tissue incision. Considering the simple strategy and outstanding performance, the PEG-PLL-Lys hydrogel is promising for clinical transformation.
Collapse
Affiliation(s)
- Changzheng Wei
- Shanghai Qisheng Biological Preparation Co., Ltd, Shanghai, 201106, China.
| | - Jialin Song
- Shanghai Qisheng Biological Preparation Co., Ltd, Shanghai, 201106, China.
| | - Haoqi Tan
- Shanghai Qisheng Biological Preparation Co., Ltd, Shanghai, 201106, China.
| |
Collapse
|
38
|
Abstract
Biopolymers are natural polymers sourced from plants and animals, which include a variety of polysaccharides and polypeptides. The inclusion of biopolymers into biomedical hydrogels is of great interest because of their inherent biochemical and biophysical properties, such as cellular adhesion, degradation, and viscoelasticity. The objective of this Review is to provide a detailed overview of the design and development of biopolymer hydrogels for biomedical applications, with an emphasis on biopolymer chemical modifications and cross-linking methods. First, the fundamentals of biopolymers and chemical conjugation methods to introduce cross-linking groups are described. Cross-linking methods to form biopolymer networks are then discussed in detail, including (i) covalent cross-linking (e.g., free radical chain polymerization, click cross-linking, cross-linking due to oxidation of phenolic groups), (ii) dynamic covalent cross-linking (e.g., Schiff base formation, disulfide formation, reversible Diels-Alder reactions), and (iii) physical cross-linking (e.g., guest-host interactions, hydrogen bonding, metal-ligand coordination, grafted biopolymers). Finally, recent advances in the use of chemically modified biopolymer hydrogels for the biofabrication of tissue scaffolds, therapeutic delivery, tissue adhesives and sealants, as well as the formation of interpenetrating network biopolymer hydrogels, are highlighted.
Collapse
Affiliation(s)
- Victoria G. Muir
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Injectable nanostructured colloidal gels resembling native nucleus pulposus as carriers of mesenchymal stem cells for the repair of degenerated intervertebral discs. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112343. [DOI: 10.1016/j.msec.2021.112343] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 01/06/2023]
|
40
|
Applications of Functionalized Hydrogels in the Regeneration of the Intervertebral Disc. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2818624. [PMID: 34458364 PMCID: PMC8397561 DOI: 10.1155/2021/2818624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023]
Abstract
Intervertebral disc degeneration (IDD) is caused by genetics, aging, and environmental factors and is one of the leading causes of low back pain. The treatment of IDD presents many challenges. Hydrogels are biomaterials that possess properties similar to those of the natural extracellular matrix and have significant potential in the field of regenerative medicine. Hydrogels with various functional qualities have recently been used to repair and regenerate diseased intervertebral discs. Here, we review the mechanisms of intervertebral disc homeostasis and degeneration and then discuss the applications of hydrogel-mediated repair and intervertebral disc regeneration. The classification of artificial hydrogels and natural hydrogels is then briefly introduced, followed by an update on the development of functional hydrogels, which include noncellular therapeutic hydrogels, cellular therapeutic hydrogel scaffolds, responsive hydrogels, and multifunctional hydrogels. The challenges faced and future developments of the hydrogels used in IDD are discussed as they further promote their clinical translation.
Collapse
|
41
|
Physicochemical and pharmacological investigations of polyvinylpyrrolidone - tetrahydroxyborate hydrogel containing the local anesthetic lidocaine. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
42
|
Nawaz HA, Schröck K, Schmid M, Krieghoff J, Maqsood I, Kascholke C, Kohn-Polster C, Schulz-Siegmund M, Hacker MC. Injectable oligomer-cross-linked gelatine hydrogels via anhydride-amine-conjugation. J Mater Chem B 2021; 9:2295-2307. [PMID: 33616150 DOI: 10.1039/d0tb02861d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Injectable gelatine-based hydrogels are valuable tools for drug and cell delivery due to their extracellular matrix-like properties that can be adjusted by the degree of cross-linking. We have established anhydride-containing oligomers for the cross-linking of gelatine via anhydride-amine-conjugation. So far, this conversion required conditions not compatible with cell encapsulation or in vivo injection. In order to overcome this limitation, we developed an array of quarter-oligomers varying in comonomer composition and contents of reactive anhydride units reactive towards amine groups under physiological conditions. The oligomers were of low molecular weight (Mn < 5 kDa) with a high degree of chemically intact anhydrides. Chemical comonomer composition was determined by 1H-NMR. Dissolutions experiments confirmed improved hydrophilicity of the synthesized oligomers over our established compositions. Injectable formulations are described utilizing cytocompatible concentrations of constituent materials and proton-scavenging base. Degree of cross-linking and stiffness of injectable hydrogels were controlled by composition. The gels hold promise as injectable drug or cell carrier and as bioink.
Collapse
Affiliation(s)
- Hafiz Awais Nawaz
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany and Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Abdul Qadir Jillani road, Lahore, Pakistan
| | - Kathleen Schröck
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Maximilian Schmid
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Jan Krieghoff
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Iram Maqsood
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Christian Kascholke
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Caroline Kohn-Polster
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Michaela Schulz-Siegmund
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany
| | - Michael C Hacker
- Institute of Pharmacy, Pharmaceutical Technology, Leipzig University, Eilenburger Straße 15 a, 04317 Leipzig, Germany and Institute of Pharmaceutics and Biopharmaceutics, Heinrich-Heine-Universität, Universitätsstraße 1, Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
43
|
Li Z, Cao H, Xu Y, Li X, Han X, Fan Y, Jiang Q, Sun Y, Zhang X. Bioinspired polysaccharide hybrid hydrogel promoted recruitment and chondrogenic differentiation of bone marrow mesenchymal stem cells. Carbohydr Polym 2021; 267:118224. [PMID: 34119177 DOI: 10.1016/j.carbpol.2021.118224] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/13/2021] [Accepted: 05/16/2021] [Indexed: 01/05/2023]
Abstract
Cartilage regeneration by biomimetic cartilage matrix with synchronously recruited stem cells was one of ideal strategies. Inspired by catechol for proteins adhesion, dopamine modified polysaccharide hybrid hydrogel (HD-C) was prepared by integrating collagen I (Col I) and hyaluronic acid derivatives (HA-DN) with sulfhydryl modified polysaccharide hybrid hydrogel (HS-C) as control. Because of double-crosslinking architecture, HD-C hydrogel was endowed with a more compact pore structure, higher mechanical properties and water retention ability in comparison with those of HS-C hydrogel. Meanwhile, it significantly promoted the proliferation and spread of rabbit bone marrow stem cells (rBMSCs), and accelerated cartilaginous matrix secretion. RT-PCR results also verified higher related gene expression of chondrogenesis (Sox 9, Agg and Col II). Moreover, HD-C hydrogel could enhance the enrichment and migration of rBMSCs in vitro by potential functional protein adsorption mechanisms, and this phenomenon was further confirmed by more rBMSCs migration in short-term joint implantation experiments in vivo.
Collapse
Affiliation(s)
- Zhulian Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Hongfu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Yang Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Xing Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - XiaoWen Han
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Qing Jiang
- College of Materials Science and Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China.
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| |
Collapse
|
44
|
Liao Z, Li S, Lu S, Liu H, Li G, Ma L, Luo R, Ke W, Wang B, Xiang Q, Song Y, Feng X, Zhang Y, Wu X, Hua W, Yang C. Metformin facilitates mesenchymal stem cell-derived extracellular nanovesicles release and optimizes therapeutic efficacy in intervertebral disc degeneration. Biomaterials 2021; 274:120850. [PMID: 33984637 DOI: 10.1016/j.biomaterials.2021.120850] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are extracellular nanovesicles that deliver diverse cargoes to the cell and participate in cell communication. Mesenchymal stem cell (MSCs)-derived EVs are considered a therapeutic approach in musculoskeletal degenerative diseases, including intervertebral disc degeneration. However, limited production yield and unstable quality have impeded the clinical application of EVs. In the present study, it is indicated that metformin promotes EVs release and alters the protein profile of EVs. Metformin enhances EVs production via an autophagy-related pathway, concomitantly with the phosphorylation of synaptosome-associated protein 29. More than quantity, quality of MSCs-derived EVs is influenced by metformin treatment. Proteomics analysis reveals that metformin increases the protein content of EVs involved in cell growth. It is shown that EVs derived from metformin-treated MSCs ameliorate intervertebral disc cells senescence in vitro and in vivo. Collectively, these findings demonstrate the great promise of metformin in EVs-based intervertebral disc regeneration.
Collapse
Affiliation(s)
- Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Saideng Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rongjin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wencan Ke
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bingjin Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qian Xiang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yukun Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
45
|
Dhand AP, Galarraga JH, Burdick JA. Enhancing Biopolymer Hydrogel Functionality through Interpenetrating Networks. Trends Biotechnol 2021; 39:519-538. [PMID: 32950262 PMCID: PMC7960570 DOI: 10.1016/j.tibtech.2020.08.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 01/22/2023]
Abstract
Traditional hydrogels are strong candidates for biomedical applications; however, they may suffer from drawbacks such as weak mechanics, static properties, and an inability to fully replicate aspects of the cellular microenvironment. These challenges can be addressed through the incorporation of second networks to form interpenetrating polymer network (IPN) hydrogels. The objective of this review is to establish clear trends on the enhanced functionality achieved by incorporating secondary networks into traditional, biopolymer-based hydrogels. These include mechanical reinforcement, 'smart' systems that respond to external stimuli, and the ability to tune cell-material interactions. Through attention to network structure and chemistry, IPN hydrogels may advance to meet challenging criteria for a wide range of biomedical fields.
Collapse
Affiliation(s)
- Abhishek P Dhand
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan H Galarraga
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Zheng K, Du D. Recent advances of hydrogel-based biomaterials for intervertebral disc tissue treatment: A literature review. J Tissue Eng Regen Med 2021; 15:299-321. [PMID: 33660950 DOI: 10.1002/term.3172] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
Low back pain is an increasingly prevalent symptom mainly associated with intervertebral disc (IVD) degeneration. It is highly correlated with aging, as the nucleus pulposus (NP) dehydrates and annulus fibrosus fissure formatting, which finally results in the IVD herniation and related clinical symptoms. Hydrogels have been drawing increasing attention as the ideal candidates for IVD degeneration because of their unique properties such as biocompatibility, highly tunable mechanical properties, and especially the water absorption and retention ability resembling the normal NP tissue. Numerous innovative hydrogel polymers have been generated in the most recent years. This review article will first briefly describe the anatomy and pathophysiology of IVDs and current therapies with their limitations. Following that, the article introduces the hydrogel materials in the classification of their origins. Next, it reviews the recent hydrogel polymers explored for IVD regeneration and analyses what efforts have been made to overcome the existing limitations. Finally, the challenges and prospects of hydrogel-based treatments for IVD tissue are also discussed. We believe that these novel hydrogel-based strategies may shed light on new possibilities in IVD degeneration disease.
Collapse
Affiliation(s)
- Kaiwen Zheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Dajiang Du
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
47
|
Abstract
Intervertebral disc (IVD) degeneration is a leading cause of chronic low back pain (LBP) that results in serious disability and significant economic burden. IVD degeneration alters the disc structure and spine biomechanics, resulting in subsequent structural changes throughout the spine. Currently, treatments of chronic LBP due to IVD degeneration include conservative treatments, such as pain medication and physiotherapy, and surgical treatments, such as removal of herniated disc without or with spinal fusion. However, none of these treatments can completely restore a degenerated disc and its function. Thus, although the exact pathogenesis of disc degeneration remains unclear, there are studies examining the effectiveness of biological approaches, such as growth factor injection, gene therapy, and cell transplantation, in promoting IVD regeneration. Furthermore, tissue engineering using a combination of cell transplantation and biomaterials has emerged as a promising new approach for repair or restoration of degenerated discs. The main purpose of this review was to provide an overview of the current status of tissue engineering applications for IVD regenerative therapy by performing literature searches using PubMed. Significant advances in tissue engineering have opened the door to a new generation of regenerative therapies for the treatment of chronic discogenic LBP.
Collapse
|
48
|
Im W, Park SY, Goo S, Yook S, Lee HL, Yang G, Youn HJ. Incorporation of CNF with Different Charge Property into PVP Hydrogel and Its Characteristics. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:426. [PMID: 33567602 PMCID: PMC7915088 DOI: 10.3390/nano11020426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/31/2021] [Accepted: 02/05/2021] [Indexed: 01/16/2023]
Abstract
Cellulose nanofibril (CNF)-added polyvinylpyrrolidone (PVP) hydrogels were prepared using different types of CNFs and their properties were investigated. CNFs with different morphology and surface charge properties were prepared through quaternization and carboxymethylation pretreatments. The quaternized CNF exhibited the narrow and uniform width, and higher viscoelastic property compared to untreated and carboxymethylated CNF. When CNF was incorporated to PVP hydrogel, gel contents of all hydrogels were similar, irrespective of CNF addition quantity or CNF type. However, the absorptivity of the hydrogels in a swelling medium increased by adding CNF. In particular, the quaternized CNF-added PVP hydrogel exhibited the highest swelling ability. Unlike that of hydrogels with untreated and carboxymethylated CNFs, the storage modulus of PVP hydrogels after swelling significantly increased with an increase in the content of the quaternized CNF. These indicate that a PVP hydrogel with a high absorptivity and storage modulus can be prepared by incorporating the proper type of CNF.
Collapse
Affiliation(s)
- Wanhee Im
- Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (W.I.); (H.L.L.)
- R&D Institute, Moorim P&P Co., 3-36 Ubonggangyang-ro, Onsan-eup, Ulju-gun, Ulsan 45011, Korea
| | - Shin Young Park
- Department of Forest Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
| | - Sooim Goo
- Department of Agriculture, Forestry and Bioresources, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.G.); (S.Y.)
| | - Simyub Yook
- Department of Agriculture, Forestry and Bioresources, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.G.); (S.Y.)
| | - Hak Lae Lee
- Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (W.I.); (H.L.L.)
- Department of Agriculture, Forestry and Bioresources, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.G.); (S.Y.)
- State Key Laboratory of Biobased Material and Green Papermaking (Shandong Academy of Sciences), Qilu University of Technology, 3501 Daxue Rd, Changqing District, Jinan 250353, China;
| | - Guihua Yang
- State Key Laboratory of Biobased Material and Green Papermaking (Shandong Academy of Sciences), Qilu University of Technology, 3501 Daxue Rd, Changqing District, Jinan 250353, China;
| | - Hye Jung Youn
- Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (W.I.); (H.L.L.)
- Department of Agriculture, Forestry and Bioresources, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (S.G.); (S.Y.)
- State Key Laboratory of Biobased Material and Green Papermaking (Shandong Academy of Sciences), Qilu University of Technology, 3501 Daxue Rd, Changqing District, Jinan 250353, China;
| |
Collapse
|
49
|
Binch ALA, Ratcliffe LPD, Milani AH, Saunders BR, Armes SP, Hoyland JA. Site-Directed Differentiation of Human Adipose-Derived Mesenchymal Stem Cells to Nucleus Pulposus Cells Using an Injectable Hydroxyl-Functional Diblock Copolymer Worm Gel. Biomacromolecules 2021; 22:837-845. [PMID: 33470795 DOI: 10.1021/acs.biomac.0c01556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adipose-derived mesenchymal stem cells (ASCs) have been identified for their promising therapeutic potential to regenerate and repopulate the degenerate intervertebral disk (IVD), which is a major cause of lower back pain. The optimal cell delivery system remains elusive but encapsulation of cells within scaffolds is likely to offer a decisive advantage over the delivery of cells in solution by ensuring successful retention within the tissue. Herein, we evaluate the use of a fully synthetic, thermoresponsive poly(glycerol monomethacrylate)-poly(2-hydroxypropyl methacrylate) (PGMA-PHPMA) diblock copolymer worm gel that mimics the structure of hydrophilic glycosaminoglycans. The objective was to use this gel to direct differentiation of human ASCs toward a nucleus pulposus (NP) phenotype, with or without the addition of discogenic growth factors TGFβ or GDF6. Accordingly, human ASCs were incorporated into a cold, free-flowing aqueous dispersion of the diblock copolymer, gelation induced by warming to 37 °C and cell culture was conducted for 14 days with or without such growth factors to assess the expression of characteristic NP markers compared to those produced when using collagen gels. In principle, the shear-thinning nature of the biocompatible worm gel enables encapsulated human ASCs to be injected into the IVD using a 21G needle. Moreover, we find significantly higher gene expression levels of ACAN, SOX-9, KRT8, and KR18 for ASCs encapsulated within worm gels compared to collagen scaffolds, regardless of the growth factors employed. In summary, such wholly synthetic worm gels offer considerable potential as an injectable cell delivery scaffold for the treatment of degenerate disk disease by promoting the transition of ASCs toward an NP-phenotype.
Collapse
Affiliation(s)
- Abbie L A Binch
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, U.K
| | - Liam P D Ratcliffe
- Department of Chemistry, University of Sheffield Brook Hill, Sheffield S3 7HF, South Yorkshire, U.K
| | - Amir H Milani
- Department of Materials, University of Manchester, Manchester M13 9PL, U.K
| | - Brian R Saunders
- Department of Materials, University of Manchester, Manchester M13 9PL, U.K
| | - Steven P Armes
- Department of Chemistry, University of Sheffield Brook Hill, Sheffield S3 7HF, South Yorkshire, U.K
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, U.K.,NIHR Manchester Biomedical Research Centre, Central Manchester Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, U.K
| |
Collapse
|
50
|
Effect of pH variation and crosslinker absence on the gelling mechanism of high acyl gellan: Morphological, thermal and mechanical approaches. Carbohydr Polym 2021; 251:117002. [DOI: 10.1016/j.carbpol.2020.117002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022]
|