1
|
Clayton SW, Walk RE, Mpofu L, Easson GWD, Tang SY. Sex-specific divergences in the types and timing of infiltrating immune cells during the intervertebral disc acute injury response and their associations with degeneration. Osteoarthritis Cartilage 2024:S1063-4584(24)01426-2. [PMID: 39426787 DOI: 10.1016/j.joca.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVE Inadequate repair of the intervertebral disc (IVD) contributes to low back pain. Infiltrating immune cells into damaged tissues are critical mediators of repair, yet little is known about the identities, roles, and temporal regulation following IVD injury. By analyzing transcripts of immune cell markers, histopathologic analysis, immunofluorescence, and flow cytometry, we aimed to define the temporal cascade of infiltrating immune cells and their associations with IVD degeneration. METHODS Caudal IVDs from 12-week-old C57BL6/J mice were injured and monitored for 42 days post-injury. Transcriptional markers identifying myeloid, B, and T immune cells, and angiogenic factors were measured from the IVDs every 2-3 days. Histopathologic degeneration of the IVD was measured throughout. Flow cytometry and immunofluorescence were used to identify and localize cells including B, T, natural killer T (NKT) cells, monocytes, neutrophils, macrophages, eosinophils, and dendritic cells. RESULTS The injured IVD revealed distinct phases of inflammation and proliferation. Robust temporal oscillation in the myeloid and T cell transcripts was observed in females. Cd3+ T cells were more abundant in females than in males. The Cd3+Cd4-Cd8- T cells that dominate the female cascade contain rare γδ T cells. Injury-mediated degeneration was prevalent in both sexes but more severe in males. CONCLUSIONS This study defines the coordinated infiltration of immune cells in the IVD following injury. We report the discovery of γδ T cells in the female IVD, and this was associated with less severe degeneration. γδ T cells have potent anti-inflammatory roles and may suppress degeneration following IVD injury.
Collapse
Affiliation(s)
| | - Remy E Walk
- Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Mpofu
- Washington University in St. Louis, St. Louis, MO, USA
| | | | - Simon Y Tang
- Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
2
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
3
|
Tie BSH, Daly M, Zhuo S, Halligan E, Keane G, Geever J, Geever L. The Exponential Shapeshifting Response of N-Vinylcaprolactam Hydrogel Bilayers Due to Temperature Change for Potential Minimally Invasive Surgery. J Funct Biomater 2024; 15:242. [PMID: 39330218 PMCID: PMC11432818 DOI: 10.3390/jfb15090242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Poly (N-vinylcaprolactam) (PNVCL) and poly (N-isopropylacrylamide) (PNIPAm) are two popular negatively temperature-responsive hydrogels, due to their biocompatibility, softness, hydrophilicity, superabsorbency, viscoelasticity, and near-physiological lower critical solution temperature (LCST). These characteristics make them ideal for biomedical applications. When combined with other materials, hydrogel expansion induces the morphing of the assembly due to internal stress differences. Our recent developments in NVCL hydrogel, enhanced by nanoclay incorporation, have driven us to the creation of a bilayer structure to study its shapeshifting response across various temperatures. This study focused on the bending behaviour of bilayer samples composed of an active hydrogel layer and a passive non-swellable layer. Using photopolymerisation, circular discs and rectangular bilayer samples of varying sizes were fabricated. Homogeneous circular samples demonstrated that hydrogel density increased proportionally with temperature, with the swelling ratio exhibiting two distinct rates of change below and above its LCST. In bilayer samples, the volume of the passive layer influenced bending, and its optimal volume was identified. The investigation revealed that geometry affected the overall bending effect due to changes in the passive layer stiffness. Lastly, a temperature-responsive gripper capable of picking up objects several times its own weight was demonstrated, highlighting the potential of NVCL hydrogels as bioactuators for minimally invasive surgery.
Collapse
Affiliation(s)
- Billy Shu Hieng Tie
- Polymer, Recycling, Industrial, Sustainability and Manufacturing (PRISM) Centre, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Mark Daly
- Faculty of Engineering & Informatics, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Shuo Zhuo
- Polymer, Recycling, Industrial, Sustainability and Manufacturing (PRISM) Centre, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Elaine Halligan
- Polymer, Recycling, Industrial, Sustainability and Manufacturing (PRISM) Centre, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Gavin Keane
- Centre for Industrial Service & Design, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Joseph Geever
- Faculty of Engineering & Informatics, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| | - Luke Geever
- Applied Polymer Technologies Gateway, Technological University of the Shannon, Midlands Midwest, N37 HD68 Athlone, Ireland
| |
Collapse
|
4
|
Clayton SW, Sebastian A, Wilson SP, Hum NR, Walk RE, Easson GWD, Vaidya R, Broz KS, Loots GG, Tang SY. Single cell RNA sequencing reveals a shift in cell function and maturation of endogenous and infiltrating cell types in response to acute intervertebral disc injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607363. [PMID: 39149307 PMCID: PMC11326235 DOI: 10.1101/2024.08.10.607363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Intervertebral disc (IVD) degeneration contributes to disabling back pain. Degeneration can be initiated by injury and progressively leads to irreversible cell loss and loss of IVD function. Attempts to restore IVD function through cell replacement therapies have had limited success due to knowledge gaps in critical cell populations and molecular crosstalk after injury. Here, we used single cell RNA sequencing to identify the transcriptional changes of endogenous and infiltrating IVD cell populations, as well as the potential of resident mesenchymal stem cells (MSCs) for tissue repair. Control and Injured (needle puncture) tail IVDs were extracted from 12 week old female C57BL/6 mice 7 days post injury and clustering analyses, gene ontology, and pseudotime trajectory analyses were used to determine transcriptomic divergences in the cells of the injured IVD, while immunofluorescence was utilized to determine mesenchymal stem cell (MSC) localization. Clustering analysis revealed 11 distinct cell populations that were IVD tissue specific, immune, or vascular cells. Differential gene expression analysis determined that Outer Annulus Fibrosus, Neutrophils, Saa2-High MSCs, Macrophages, and Krt18+ Nucleus Pulposus (NP) cells were the major drivers of transcriptomic differences between Control and Injured cells. Gene ontology of DEGs suggested that the most upregulated biological pathways were angiogenesis and T cell related while wound healing and ECM regulation categories were downregulated. Pseudotime trajectory analyses revealed that cells were driven towards increased cell differentiation due to IVD injury in all IVD tissue clusters except for Krt18+ NP which remained in a less mature cell state. Saa2-High and Grem1-High MSCs populations drifted towards more IVD differentiated cells profiles with injury and localized distinctly within the IVD. This study strengthens the understanding of heterogeneous IVD cell populations response to injury and identifies targetable MSC populations for future IVD repair studies.
Collapse
Affiliation(s)
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore CA
| | - Stephen P Wilson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore CA
| | - Nicholas R Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore CA
| | - Remy E Walk
- Washington University in St. Louis, St. Louis MO
| | | | | | | | - Gabriela G Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore CA
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States
| | - Simon Y Tang
- Washington University in St. Louis, St. Louis MO
| |
Collapse
|
5
|
Nguyen CT, Chow SKK, Nguyen HN, Liu T, Walls A, Withey S, Liebig P, Mueller M, Thierry B, Yang CT, Huang CJ. Formation of Zwitterionic and Self-Healable Hydrogels via Amino-yne Click Chemistry for Development of Cellular Scaffold and Tumor Spheroid Phantom for MRI. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36157-36167. [PMID: 38973633 PMCID: PMC11261563 DOI: 10.1021/acsami.4c06917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
In situ-forming biocompatible hydrogels have great potential in various medical applications. Here, we introduce a pH-responsive, self-healable, and biocompatible hydrogel for cell scaffolds and the development of a tumor spheroid phantom for magnetic resonance imaging. The hydrogel (pMAD) was synthesized via amino-yne click chemistry between poly(2-methacryloyloxyethyl phosphorylcholine-co-2-aminoethylmethacrylamide) and dialkyne polyethylene glycol. Rheology analysis, compressive mechanical testing, and gravimetric analysis were employed to investigate the gelation time, mechanical properties, equilibrium swelling, and degradability of pMAD hydrogels. The reversible enamine and imine bond mechanisms leading to the sol-to-gel transition in acidic conditions (pH ≤ 5) were observed. The pMAD hydrogel demonstrated potential as a cellular scaffold, exhibiting high viability and NIH-3T3 fibroblast cell encapsulation under mild conditions (37 °C, pH 7.4). Additionally, the pMAD hydrogel also demonstrated the capability for in vitro magnetic resonance imaging of glioblastoma tumor spheroids based on the chemical exchange saturation transfer effect. Given its advantages, the pMAD hydrogel emerges as a promising material for diverse biomedical applications, including cell carriers, bioimaging, and therapeutic agent delivery.
Collapse
Affiliation(s)
- Cao Tuong
Vi Nguyen
- Department
of Chemical & Materials Engineering, National Central University, Jhong-Li, Taoyuan 320, Taiwan
| | - Steven Kwok Keung Chow
- Clinical
Research and Imaging Centre, South Australian
Health and Medical Research Institute, Adelaide 5001, Australia
| | - Hoang Nam Nguyen
- Department
of Chemical & Materials Engineering, National Central University, Jhong-Li, Taoyuan 320, Taiwan
| | - Tesi Liu
- Future
Industries Institute, University of South
Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Angela Walls
- Clinical
Research and Imaging Centre, South Australian
Health and Medical Research Institute, Adelaide 5001, Australia
| | | | | | - Marco Mueller
- Advanced
Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne 1000, Switzerland
| | - Benjamin Thierry
- Future
Industries Institute, University of South
Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Chih-Tsung Yang
- Future
Industries Institute, University of South
Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Chun-Jen Huang
- Department
of Chemical & Materials Engineering, National Central University, Jhong-Li, Taoyuan 320, Taiwan
- R&D
Center for Membrane Technology, Chung Yuan
Christian University, 200 Chung Pei Road, Chung-Li City 32023, Taiwan
| |
Collapse
|
6
|
Wan S, Aregueta Robles U, Poole-Warren L, Esrafilzadeh D. Advances in 3D tissue models for neural engineering: self-assembled versus engineered tissue models. Biomater Sci 2024; 12:3522-3549. [PMID: 38829222 DOI: 10.1039/d4bm00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Neural tissue engineering has emerged as a promising field that aims to create functional neural tissue for therapeutic applications, drug screening, and disease modelling. It is becoming evident in the literature that this goal requires development of three-dimensional (3D) constructs that can mimic the complex microenvironment of native neural tissue, including its biochemical, mechanical, physical, and electrical properties. These 3D models can be broadly classified as self-assembled models, which include spheroids, organoids, and assembloids, and engineered models, such as those based on decellularized or polymeric scaffolds. Self-assembled models offer advantages such as the ability to recapitulate neural development and disease processes in vitro, and the capacity to study the behaviour and interactions of different cell types in a more realistic environment. However, self-assembled constructs have limitations such as lack of standardised protocols, inability to control the cellular microenvironment, difficulty in controlling structural characteristics, reproducibility, scalability, and lengthy developmental timeframes. Integrating biomimetic materials and advanced manufacturing approaches to present cells with relevant biochemical, mechanical, physical, and electrical cues in a controlled tissue architecture requires alternate engineering approaches. Engineered scaffolds, and specifically 3D hydrogel-based constructs, have desirable properties, lower cost, higher reproducibility, long-term stability, and they can be rapidly tailored to mimic the native microenvironment and structure. This review explores 3D models in neural tissue engineering, with a particular focus on analysing the benefits and limitations of self-assembled organoids compared with hydrogel-based engineered 3D models. Moreover, this paper will focus on hydrogel based engineered models and probe their biomaterial components, tuneable properties, and fabrication techniques that allow them to mimic native neural tissue structures and environment. Finally, the current challenges and future research prospects of 3D neural models for both self-assembled and engineered models in neural tissue engineering will be discussed.
Collapse
Affiliation(s)
- Shuqian Wan
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Ulises Aregueta Robles
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
- Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
7
|
Rizzo A, Ajò A, Kang H, De Cola L, Jesus B. Development of a new kappa-carrageenan hydrogel system to study benthic diatom vertical movements. PLoS One 2024; 19:e0297962. [PMID: 38603710 PMCID: PMC11008860 DOI: 10.1371/journal.pone.0297962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/15/2024] [Indexed: 04/13/2024] Open
Abstract
Benthic diatom vertical movement has been investigated mainly through indirect measurements based on chlorophyll a fluorescence and spectral reflectance signals. The presence of sediment hinders direct imaging and grazers activity renders the work under controlled conditions very difficult. This study provides a tool to study diatoms movement in a 3D hydrogel matrix. Synthetic and natural hydrogels were tested to find the best 3D transparent scaffold where diatoms could grow and freely move in all directions. Polyamidoamines (PAAm) hydrogels were no-cytocompatible and hyaluronic acid (HA) only allowed diatoms to survive for 2-days. Natural hydrogels made of gelatin/Na-alginate, Na-alginate and kappa-carrageenan (KC) were cytocompatible, with KC showing the best properties for diatom growth and movement on a long term (up to 2 months). Comparing Nitzschia spathulata, Gyrosigma limosum and Navicula phyllepta growth in liquid media vs in KC gels, we found that diatoms reached a significantly higher final biomass in the hydrogel condition. Hydrogels were also useful to isolate large size diatom species e.g., Nitzschia elongata, that did not survive in suspension. Finally, we showed three ways to study diatom species-specific movement in KC hydrogels: 1) controlled species mix; 2) natural diatom assemblages with grazers; and 3) natural diatom assemblages without grazers. With our system, single diatoms could be imaged, identified, and counted. In addition, different stimuli, e.g., light intensity and light composition can be applied and their effects on movement and physiology studied without being masked by sediment or impaired by meiofauna.
Collapse
Affiliation(s)
- Arianna Rizzo
- Institut des Substances et Organismes de la Mer–ISOMer UR 2160, Faculté des Sciences et des Techniques, Nantes University, Nantes, France
| | - Alessandro Ajò
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milano, Italy
- Pharmaceutical Science Department, University of Milan, Milan, Italy
| | - Huixuan Kang
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milano, Italy
- Pharmaceutical Science Department, University of Milan, Milan, Italy
| | - Luisa De Cola
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milano, Italy
- Pharmaceutical Science Department, University of Milan, Milan, Italy
| | - Bruno Jesus
- Institut des Substances et Organismes de la Mer–ISOMer UR 2160, Faculté des Sciences et des Techniques, Nantes University, Nantes, France
| |
Collapse
|
8
|
Clayton SW, Walk RE, Mpofu L, Easson GW, Tang SY. Analysis of Infiltrating Immune Cells Following Intervertebral Disc Injury Reveals Recruitment of Gamma-Delta ( γδ) T cells in Female Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582950. [PMID: 38464124 PMCID: PMC10925253 DOI: 10.1101/2024.03.01.582950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Inadequate repair of injured intervertebral discs (IVD) leads to degeneration and contributes to low back pain. Infiltrating immune cells into damaged musculoskeletal tissues are critical mediators of repair, yet little is known about their identities, roles, and temporal regulation following IVD injury. By analyzing longitudinal changes in gene expression, tissue morphology, and the dynamics of infiltrating immune cells following injury, we characterize sex-specific differences in immune cell populations and identify the involvement of previously unreported immune cell types, γδ and NKT cells. Cd3+Cd4-Cd8- T cells are the largest infiltrating lymphocyte population with injury, and we identified the presence of γδ T cells in this population in female mice specifically, and NKT cells in males. Injury-mediated IVD degeneration was prevalent in both sexes, but more severe in males. Sex-specific degeneration may be associated with the differential immune response since γδ T cells have potent anti-inflammatory roles and may mediate IVD repair.
Collapse
Affiliation(s)
| | - Remy E. Walk
- Washington University in St. Louis, St. Louis, MO
| | - Laura Mpofu
- Washington University in St. Louis, St. Louis, MO
| | | | | |
Collapse
|
9
|
Wu J, Yun Z, Song W, Yu T, Xue W, Liu Q, Sun X. Highly oriented hydrogels for tissue regeneration: design strategies, cellular mechanisms, and biomedical applications. Theranostics 2024; 14:1982-2035. [PMID: 38505623 PMCID: PMC10945336 DOI: 10.7150/thno.89493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/19/2024] [Indexed: 03/21/2024] Open
Abstract
Many human tissues exhibit a highly oriented architecture that confers them with distinct mechanical properties, enabling adaptation to diverse and challenging environments. Hydrogels, with their water-rich "soft and wet" structure, have emerged as promising biomimetic materials in tissue engineering for repairing and replacing damaged tissues and organs. Highly oriented hydrogels can especially emulate the structural orientation found in human tissue, exhibiting unique physiological functions and properties absent in traditional homogeneous isotropic hydrogels. The design and preparation of highly oriented hydrogels involve strategies like including hydrogels with highly oriented nanofillers, polymer-chain networks, void channels, and microfabricated structures. Understanding the specific mechanism of action of how these highly oriented hydrogels affect cell behavior and their biological applications for repairing highly oriented tissues such as the cornea, skin, skeletal muscle, tendon, ligament, cartilage, bone, blood vessels, heart, etc., requires further exploration and generalization. Therefore, this review aims to fill that gap by focusing on the design strategy of highly oriented hydrogels and their application in the field of tissue engineering. Furthermore, we provide a detailed discussion on the application of highly oriented hydrogels in various tissues and organs and the mechanisms through which highly oriented structures influence cell behavior.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhihe Yun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wenlong Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130023, China
| | - Tao Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wu Xue
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qinyi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xinzhi Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
10
|
Riahinezhad H, Amsden BG. In situ forming, mechanically resilient hydrogels prepared from 4a-[PEG- b-PTMC-Ac] and thiolated chondroitin sulfate for nucleus pulposus cell delivery. J Mater Chem B 2024; 12:1257-1270. [PMID: 38167961 DOI: 10.1039/d3tb02574h] [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: 01/05/2024]
Abstract
Intervertebral disk degeneration (IVDD) is a common condition that causes severe back pain and affects patients' mobility and life quality considerably. IVDD originates within the central region of the disk called the nucleus pulposus (NP). Removing the damaged tissue and replacing it with NP cells (NPCs) delivered within an in situ forming hydrogel is a promising treatment approach. Herein we describe a hydrogel formulation based on 4-arm [poly(ethylene glycol)-b-poly(trimethylene carbonate)-acrylate] (4a[PEG-b-PTMC-Ac]) crosslinked with thiolated chondroitin sulfate via Michael-type reaction for this purpose. A library of hydrogels based on 15 kDa 4a-[PEG] with PTMC blocks of varying molecular weight were prepared and characterized. The instantaneous moduli of the hydrogels were adjustable from 24 to 150 kPa depending on the length of the PTMC block and the polymer volume fraction. The influence of each of these parameters was effectively explained using both scaling or mean field theories of polyelectrolyte hydrogels. The hydrogels were resistant to cyclic compressive loading and degraded gradually over 70 days in vitro. A hydrogel formulation with an instantaneous modulus at the high end of the range of values reported for human NP tissue was chosen to assess the ability of these hydrogels for delivering NPCs. The prepolymer solution was injectable and formed a hydrogel within 30 minutes at 37 °C. Bovine NPCs were encapsulated within this hydrogel with high viability and proliferated throughout a 28 day, hypoxic culture period. The encapsulated NPCs formed clusters and deposited collagen type II but no collagen type I within the hydrogels. Despite an initial gradual decrease, a steady-state modulus was reached at the end of the 28 day culture period that was within the range reported for healthy human NP tissue. This in situ forming hydrogel formulation is a promising approach and with further development could be a viable clinical treatment for IVDD.
Collapse
Affiliation(s)
- Hossein Riahinezhad
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada.
| | - Brian G Amsden
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
11
|
Yang S, Jing S, Wang S, Jia F. From drugs to biomaterials: a review of emerging therapeutic strategies for intervertebral disc inflammation. Front Cell Infect Microbiol 2024; 14:1303645. [PMID: 38352058 PMCID: PMC10861683 DOI: 10.3389/fcimb.2024.1303645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Chronic low back pain (LBP) is an increasingly prevalent issue, especially among aging populations. A major underlying cause of LBP is intervertebral disc degeneration (IDD), often triggered by intervertebral disc (IVD) inflammation. Inflammation of the IVD is divided into Septic and Aseptic inflammation. Conservative therapy and surgical treatment often fail to address the root cause of IDD. Recent advances in the treatment of IVD infection and inflammation range from antibiotics and small-molecule drugs to cellular therapies, biological agents, and innovative biomaterials. This review sheds light on the complex mechanisms of IVD inflammation and physiological and biochemical processes of IDD. Furthermore, it provides an overview of recent research developments in this area, intending to identify novel therapeutic targets and guide future clinical strategies for effectively treating IVD-related conditions.
Collapse
Affiliation(s)
- Shuhan Yang
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Shaoze Jing
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Shanxi Wang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Fajing Jia
- Department of General Practice, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
12
|
Hidalgo-Alvarez V, Madl CM. Leveraging Biomaterial Platforms to Study Aging-Related Neural and Muscular Degeneration. Biomolecules 2024; 14:69. [PMID: 38254669 PMCID: PMC10813704 DOI: 10.3390/biom14010069] [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: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Aging is a complex multifactorial process that results in tissue function impairment across the whole organism. One of the common consequences of this process is the loss of muscle mass and the associated decline in muscle function, known as sarcopenia. Aging also presents with an increased risk of developing other pathological conditions such as neurodegeneration. Muscular and neuronal degeneration cause mobility issues and cognitive impairment, hence having a major impact on the quality of life of the older population. The development of novel therapies that can ameliorate the effects of aging is currently hindered by our limited knowledge of the underlying mechanisms and the use of models that fail to recapitulate the structure and composition of the cell microenvironment. The emergence of bioengineering techniques based on the use of biomimetic materials and biofabrication methods has opened the possibility of generating 3D models of muscular and nervous tissues that better mimic the native extracellular matrix. These platforms are particularly advantageous for drug testing and mechanistic studies. In this review, we discuss the developments made in the creation of 3D models of aging-related neuronal and muscular degeneration and we provide a perspective on the future directions for the field.
Collapse
Affiliation(s)
| | - Christopher M. Madl
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
13
|
Zhou D, Liu H, Zheng Z, Wu D. Design principles in mechanically adaptable biomaterials for repairing annulus fibrosus rupture: A review. Bioact Mater 2024; 31:422-439. [PMID: 37692911 PMCID: PMC10485601 DOI: 10.1016/j.bioactmat.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/25/2023] [Accepted: 08/12/2023] [Indexed: 09/12/2023] Open
Abstract
Annulus fibrosus (AF) plays a crucial role in the biomechanical loading of intervertebral disc (IVD). AF is difficult to self-heal when the annulus tears develop, because AF has a unique intricate structure and biologic milieu in vivo. Tissue engineering is promising for repairing AF rupture, but construction of suitable mechanical matching devices or scaffolds is still a grand challenge. To deeply know the varied forces involved in the movement of the native annulus is highly beneficial for designing biomimetic scaffolds to recreate the AF function. In this review, we overview six freedom degrees of forces and adhesion strength on AF tissue. Then, we summarize the mechanical modalities to simulate related forces on AF and to assess the characteristics of biomaterials. We finally outline some current advanced techniques to develop mechanically adaptable biomaterials for AF rupture repair.
Collapse
Affiliation(s)
- Dan Zhou
- 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
|
14
|
Orieshyna A, Puetzer JL, Amdursky N. Proton Transport Across Collagen Fibrils and Scaffolds: The Role of Hydroxyproline. Biomacromolecules 2023; 24:4653-4662. [PMID: 37656903 DOI: 10.1021/acs.biomac.3c00326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Collagen is one of the most studied proteins due to its fundamental role in creating fibrillar structures and supporting tissues in our bodies. Accordingly, collagen is also one of the most used proteins for making tissue-engineered scaffolds for various types of tissues. To date, the high abundance of hydroxyproline (Hyp) within collagen is commonly ascribed to the structure and stability of collagen. Here, we hypothesize a new role for the presence of Hyp within collagen, which is to support proton transport (PT) across collagen fibrils. For this purpose, we explore here three different collagen-based hydrogels: the first is prepared by the self-assembly of natural collagen fibrils, and the second and third are based on covalently linking between collagen via either a self-coupling method or with an additional cross-linker. Following the formation of the hydrogel, we introduce here a two-step reaction, involving (1) attaching methanesulfonyl to the -OH group of Hyp, followed by (2) removing the methanesulfonyl, thus reverting Hyp to proline (Pro). We explore the PT efficiency at each step of the reaction using electrical measurements and show that adding the methanesulfonyl group vastly enhances PT, while reverting Hyp to Pro significantly reduces PT efficiency (compared with the initial point) with different efficiencies for the various collagen-based hydrogels. The role of Hyp in supporting the PT can assist in our understanding of the physiological roles of collagen. Furthermore, the capacity to modulate conductivity across collagen is very important to the use of collagen in regenerative medicine.
Collapse
Affiliation(s)
- Anna Orieshyna
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Nadav Amdursky
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
15
|
Chen R, Liu Z, Cui T, Zhang X, Wang CF, Li GX, Wang G, Chen S. HE@PCL/PCE Gel-Nanofiber Dressing with Robust Self-Adhesion toward High Wound-Healing Rate via Microfluidic Electrospinning Technology. ACS APPLIED MATERIALS & INTERFACES 2023; 15:46322-46332. [PMID: 37748017 DOI: 10.1021/acsami.3c09713] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Hydrogels have attracted increasing attention in the biomedical field due to their similarity in structure and composition to natural extracellular matrices. However, they have been greatly limited by their low mechanical strength and self-adhesion for further application. Here, a gel-nanofiber material is designed for wound healing, which synergistically combines the benefits of hydrogels and nanofibers and can overcome the bottleneck of poor mechanical strength and self-adhesion in hydrogels and inadequate healing environment created by nanofibers. First, a nanofiber scaffold composed of polycaprolactone/poly(citric acid)-ε-lysine (PCL/PCE) nanofibers is fabricated via a new strategy of microfluidic electrospinning, which could provide a base for hyaluronic acid-polylysine (HE) gel growth on nanofibers. The prepared HE@PCL/PCE gel-nanofiber possesses high tensile strength (24.15 ± 1.67 MPa), excellent air permeability (656 m3/m2 h kPa), outstanding self-adhesion property, and positive hydrophilicity. More importantly, the prepared gel-nanofiber dressing shows good cytocompatibility and antibacterial properties, achieving a high wound-healing rate (92.48%) and 4.685 mm granulation growth thickness within 12 days. This material may open a promising avenue for accelerating wound healing and tissue regeneration, providing potential applications in clinical medicine.
Collapse
Affiliation(s)
- Rong Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu Key Laboratory of Fine Chemicals and Functional Polymer Materials, Nanjing Tech University, No. 5 Xin Mofan Road, Nanjing 210009, P. R. China
| | - Zhiting Liu
- Department of General Surgery, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, 305 East Zhongshan Road, Nanjing 210002, China
| | - Tingting Cui
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu Key Laboratory of Fine Chemicals and Functional Polymer Materials, Nanjing Tech University, No. 5 Xin Mofan Road, Nanjing 210009, P. R. China
| | - Xiaoying Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu Key Laboratory of Fine Chemicals and Functional Polymer Materials, Nanjing Tech University, No. 5 Xin Mofan Road, Nanjing 210009, P. R. China
| | - Cai-Feng Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu Key Laboratory of Fine Chemicals and Functional Polymer Materials, Nanjing Tech University, No. 5 Xin Mofan Road, Nanjing 210009, P. R. China
| | - Guo-Xing Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu Key Laboratory of Fine Chemicals and Functional Polymer Materials, Nanjing Tech University, No. 5 Xin Mofan Road, Nanjing 210009, P. R. China
| | - Gefei Wang
- Department of General Surgery, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, 305 East Zhongshan Road, Nanjing 210002, China
| | - Su Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Jiangsu Key Laboratory of Fine Chemicals and Functional Polymer Materials, Nanjing Tech University, No. 5 Xin Mofan Road, Nanjing 210009, P. R. China
| |
Collapse
|
16
|
Belgodere JA, Lassiter HR, Robinson JT, Hamel KM, Rogers EL, Mohiuddin OA, Zhang L, Wu X, Gimble JM, Frazier TP, Monroe WT, Sanchez CG. Biomechanical and Biological Characterization of XGel, a Human-Derived Hydrogel for Stem Cell Expansion and Tissue Engineering. Adv Biol (Weinh) 2023; 7:e2200332. [PMID: 37236203 DOI: 10.1002/adbi.202200332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/23/2023] [Indexed: 05/28/2023]
Abstract
Hydrogels are 3D scaffolds used as alternatives to in vivo models for disease modeling and delivery of cells and drugs. Existing hydrogel classifications include synthetic, recombinant, chemically defined, plant- or animal-based, and tissue-derived matrices. There is a need for materials that can support both human tissue modeling and clinically relevant applications requiring stiffness tunability. Human-derived hydrogels are not only clinically relevant, but they also minimize the use of animal models for pre-clinical studies. This study aims to characterize XGel, a new human-derived hydrogel as an alternative to current murine-derived and synthetic recombinant hydrogels that features unique physiochemical, biochemical, and biological properties that support adipocyte and bone differentiation. Rheology studies determine the viscosity, stiffness, and gelation features of XGel. Quantitative studies for quality control support consistency in the protein content between lots. Proteomics studies reveal that XGel is predominantly composed of extracellular matrix proteins, including fibrillin, collagens I-VI, and fibronectin. Electron microscopy of the hydrogel provides phenotypic characteristics in terms of porosity and fiber size. The hydrogel demonstrates biocompatibility as a coating material and as a 3D scaffold for the growth of multiple cell types. The results provide insight into the biological compatibility of this human-derived hydrogel for tissue engineering.
Collapse
Affiliation(s)
- Jorge A Belgodere
- Department of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, LA, 70803, USA
| | | | | | | | | | - Omair A Mohiuddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Liwen Zhang
- Campus Chemical Instrument Center Proteomics Shared Resources, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA, 70148, USA
| | | | | | - William T Monroe
- Department of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, LA, 70803, USA
| | | |
Collapse
|
17
|
Li C, Liu Y, Weng T, Yang M, Wang X, Chai W. Fabrication of Injectable Kartogenin-Conjugated Composite Hydrogel with a Sustained Drug Release for Cartilage Repair. Pharmaceutics 2023; 15:1949. [PMID: 37514135 PMCID: PMC10385945 DOI: 10.3390/pharmaceutics15071949] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Cartilage tissue engineering has attracted great attention in defect repair and regeneration. The utilization of bioactive scaffolds to effectively regulate the phenotype and proliferation of chondrocytes has become an elemental means for cartilage tissue regeneration. On account of the simultaneous requirement of mechanical and biological performances for tissue-engineered scaffolds, in this work we prepared a naturally derived hydrogel composed of a bioactive kartogenin (KGN)-linked chitosan (CS-KGN) and an aldehyde-modified oxidized alginate (OSA) via the highly efficient Schiff base reaction and multifarious physical interactions in mild conditions. On the basis of the rigid backbones and excellent biocompatibility of these two natural polysaccharides, the composite hydrogel demonstrated favorable morphology, easy injectability, good mechanical strength and tissue adhesiveness, low swelling ratio, long-term sustainable KGN release, and facilitated bone marrow mesenchymal stem cell activity, which could simultaneously provide the mechanical and biological supports to promote chondrogenic differentiation and repair the articular cartilage defects. Therefore, we believe this work can offer a designable consideration and potential alternative candidate for cartilage and other soft tissue implants.
Collapse
Affiliation(s)
- Chao Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yubo Liu
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Tujun Weng
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Muyuan Yang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei Chai
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| |
Collapse
|
18
|
Jarrah RM, Potes MDA, Vitija X, Durrani S, Ghaith AK, Mualem W, Zamanian C, Bhandarkar AR, Bydon M. Alginate hydrogels: A potential tissue engineering intervention for intervertebral disc degeneration. J Clin Neurosci 2023; 113:32-37. [PMID: 37159956 DOI: 10.1016/j.jocn.2023.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 05/11/2023]
Abstract
Intervertebral disc (IVD) degeneration is a major cause of low back pain and disability, affecting millions of people worldwide. Current treatments for IVD degeneration are limited to invasive surgery or pain management. Recently, there has been increasing interest in the use of biomaterials, such as alginate hydrogels, for the treatment of IVD degeneration. Alginate hydrogels are an example of such a biomaterial that is biocompatible and can be tailored to mimic the native extracellular matrix of the IVD. Derived from alginate, a naturally derived polysaccharide from brown seaweed that can be transformed into a gelatinous solution, alginate hydrogels are emerging in the field of tissue engineering. They can be used to deliver therapeutic agents, such as growth factors or cells, to the site of injury, providing a localized and sustained release that may enhance treatment outcomes. This paper provides an overview on the use of alginate hydrogels for the treatment of IVD degeneration. We discuss the properties of alginate hydrogels and their potential applications for IVD regeneration, including the mechanism against IVD degeneration. We also highlight the research outcomes to date along with the challenges and limitations of using alginate hydrogels for IVD regeneration, including their mechanical properties, biocompatibility, and surgical compatibility. Overall, this review paper aims to provide a comprehensive overview of the current research on alginate hydrogels for IVD degeneration and to identify future directions for research in this area.
Collapse
Affiliation(s)
- Ryan M Jarrah
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Maria D Astudillo Potes
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Xheneta Vitija
- Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA; College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Sulaman Durrani
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Abdul Karim Ghaith
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - William Mualem
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Cameron Zamanian
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Archis R Bhandarkar
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Mohamad Bydon
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
19
|
Koo YW, Lim CS, Darai A, Lee J, Kim W, Han I, Kim GH. Shape-memory collagen scaffold combined with hyaluronic acid for repairing intervertebral disc. Biomater Res 2023; 27:26. [PMID: 36991502 DOI: 10.1186/s40824-023-00368-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a common cause of chronic low back pain (LBP) and a socioeconomic burden worldwide. Conservative therapies and surgical treatments provide only symptomatic pain relief without promoting intervertebral disc (IVD) regeneration. Therefore, the clinical demand for disc regenerative therapies for disc repair is high. METHODS In this study, we used a rat tail nucleotomy model to develop mechanically stable collagen-cryogel and fibrillated collagen with shape-memory for use in minimally invasive surgery for effective treatment of IVDD. The collagen was loaded with hyaluronic acid (HA) into a rat tail nucleotomy model. RESULTS The shape-memory collagen structures exhibited outstanding chondrogenic activities, having completely similar physical properties to those of a typical shape-memory alginate construct in terms of water absorption, compressive properties, and shape-memorability behavior. The treatment of rat tail nucleotomy model with shape-memory collagen-cryogel/HA alleviated mechanical allodynia, maintained a higher concentration of water content, and preserved the disc structure by restoring the matrix proteins. CONCLUSION According to these results, the collagen-based structure could effectively repair and maintain the IVD matrix better than the controls, including HA only and shape-memory alginate with HA.
Collapse
Affiliation(s)
- Young Won Koo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Chang Su Lim
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-Si, Gyeonggi-Do, 13496, Republic of Korea
| | - Anjani Darai
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-Si, Gyeonggi-Do, 13496, Republic of Korea
| | - JiUn Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Wonjin Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-Si, Gyeonggi-Do, 13496, Republic of Korea.
| | - Geun Hyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
- Department of Biophysics, Institute of Quantum Biophysics , Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
20
|
Khalid S, Ekram S, Ramzan F, Salim A, Khan I. Co-regulation of Sox9 and TGFβ1 transcription factors in mesenchymal stem cells regenerated the intervertebral disc degeneration. Front Med (Lausanne) 2023; 10:1127303. [PMID: 37007782 PMCID: PMC10063891 DOI: 10.3389/fmed.2023.1127303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
BackgroundIntervertebral disc (IVD) shows aging and degenerative changes earlier than any other body connective tissue. Its repair and regeneration provide a considerable challenge in regenerative medicine due to its high degree of infrastructure and mechanical complexity. Mesenchymal stem cells, due to their tissue resurfacing potential, represent many explanatory pathways to regenerate a tissue breakdown.MethodsThis study was undertaken to evaluate the co-regulation of Sox9 and TGFβ1 in differentiating human umbilical cord mesenchymal stem cells (hUC-MSC) into chondrocytes. The combinatorial impact of Sox9 and TGFβ1 on hUC-MSCs was examined in vitro by gene expression and immunocytochemical staining. In in vivo, an animal model of IVD degeneration was established under a fluoroscopic guided system through needle puncture of the caudal disc. Normal and transfected MSCs were transplanted. Oxidative stress, pain, and inflammatory markers were evaluated by qPCR. Disc height index (DHI), water content, and gag content were analyzed. Histological examinations were performed to evaluate the degree of regeneration.ResultshUC-MSC transfected with Sox9+TGFβ1 showed a noticeable morphological appearance of a chondrocyte, and highly expressed chondrogenic markers (aggrecan, Sox9, TGFβ1, TGFβ2, and type II collagens) after transfection. Histological observation demonstrated that cartilage regeneration, extracellular matrix synthesis, and collagen remodeling were significant upon staining with H&E, Alcian blue, and Masson's trichrome stain on day 14. Additionally, oxidative stress, pain, and inflammatory markers were positively downregulated in the animals transplanted with Sox9 and TGFβ1 transfected MSCs.ConclusionThese findings indicate that the combinatorial effect of Sox9 and TGFβ1 substantially accelerates the chondrogenesis in hUC-MSCs. Cartilage regeneration and matrix synthesis were significantly enhanced. Therefore, a synergistic effect of Sox9 and TGFβ1 could be an immense therapeutic combination in the tissue engineering of cartilaginous joint bio-prostheses and a novel candidate for cartilage stabilization.
Collapse
|
21
|
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
|
22
|
Properties and Printability of the Synthesized Hydrogel Based on GelMA. Int J Mol Sci 2023; 24:ijms24032121. [PMID: 36768446 PMCID: PMC9917366 DOI: 10.3390/ijms24032121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Gelatin methacryloyl (GelMA) has recently attracted increasing attention. Unlike other hydrogels, it allows for the adjustment of the mechanical properties using such factors as degree of functionalization, concentration, and photocrosslinking parameters. In this study, GelMA with a high degree of substitution (82.75 ± 7.09%) was synthesized, and its suitability for extrusion printing, cytocompatibility, and biocompatibility was studied. Satisfactory printing quality was demonstrated with the 15% concentration hydrogel. The high degree of functionalization led to a decrease in the ability of human adipose-derived stem cells (ADSCs) to adhere to the GelMA surface. During the first 3 days after sowing, proliferation was observed. Degradation in animals after subcutaneous implantation was slowed down.
Collapse
|
23
|
Arul MR, Zhang C, Alahmadi I, Moss IL, Banasavadi-Siddegowda YK, Abdulmalik S, Illien-Junger S, Kumbar SG. Novel Injectable Fluorescent Polymeric Nanocarriers for Intervertebral Disc Application. J Funct Biomater 2023; 14:52. [PMID: 36826851 PMCID: PMC9961171 DOI: 10.3390/jfb14020052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Damage to intervertebral discs (IVD) can lead to chronic pain and disability, and no current treatments can fully restore their function. Some non-surgical treatments have shown promise; however, these approaches are generally limited by burst release and poor localization of diverse molecules. In this proof-of-concept study, we developed a nanoparticle (NP) delivery system to efficiently deliver high- and low-solubility drug molecules. Nanoparticles of cellulose acetate and polycaprolactone-polyethylene glycol conjugated with 1-oxo-1H-pyrido [2,1-b][1,3]benzoxazole-3-carboxylic acid (PBC), a novel fluorescent dye, were prepared by the oil-in-water emulsion. Two drugs, a water insoluble indomethacin (IND) and a water soluble 4-aminopyridine (4-AP), were used to study their release patterns. Electron microscopy confirmed the spherical nature and rough surface of nanoparticles. The particle size analysis revealed a hydrodynamic radius ranging ~150-162 nm based on dynamic light scattering. Zeta potential increased with PBC conjugation implying their enhanced stability. IND encapsulation efficiency was almost 3-fold higher than 4-AP, with release lasting up to 4 days, signifying enhanced solubility, while the release of 4-AP continued for up to 7 days. Nanoparticles and their drug formulations did not show any apparent cytotoxicity and were taken up by human IVD nucleus pulposus cells. When injected into coccygeal mouse IVDs in vivo, the nanoparticles remained within the nucleus pulposus cells and the injection site of the nucleus pulposus and annulus fibrosus of the IVD. These fluorescent nano-formulations may serve as a platform technology to deliver therapeutic agents to IVDs and other tissues that require localized drug injections.
Collapse
Affiliation(s)
- Michael R. Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | - Changli Zhang
- Department of Orthopedic Surgery, Emory University, Atlanta, GA 30308, USA
| | - Ibtihal Alahmadi
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Isaac L. Moss
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | | | - Sama Abdulmalik
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | | | - Sangamesh G. Kumbar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
24
|
Li W, Zhou P, Yan B, Qi M, Chen Y, Shang L, Guan J, Zhang L, Mao Y. Disc regeneration by injectable fucoidan-methacrylated dextran hydrogels through mechanical transduction and macrophage immunomodulation. J Tissue Eng 2023; 14:20417314231180050. [PMID: 37427012 PMCID: PMC10328174 DOI: 10.1177/20417314231180050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/19/2023] [Indexed: 07/11/2023] Open
Abstract
Modulating a favorable inflammatory microenvironment that facilitates the recovery of degenerated discs is a key strategy in the treatment of intervertebral disc (IVD) degeneration (IDD). More interestingly, well-mechanized tissue-engineered scaffolds have been proven in recent years to be capable of sensing mechanical transduction to enhance the proliferation and activation of nucleus pulposus cells (NPC) and have demonstrated an increased potential in the treatment and recovery of degenerative discs. Additionally, existing surgical procedures may not be suitable for IDD treatment, warranting the requirement of new regenerative therapies for the restoration of disc structure and function. In this study, a light-sensitive injectable polysaccharide composite hydrogel with excellent mechanical properties was prepared using dextrose methacrylate (DexMA) and fucoidan with inflammation-modulating properties. Through numerous in vivo experiments, it was shown that the co-culture of this composite hydrogel with interleukin-1β-stimulated NPCs was able to promote cell proliferation whilst preventing inflammation. Additionally, activation of the caveolin1-yes-associated protein (CAV1-YAP) mechanotransduction axis promoted extracellular matrix (ECM) metabolism and thus jointly promoted IVD regeneration. After injection into an IDD rat model, the composite hydrogel inhibited the local inflammatory response by inducing macrophage M2 polarization and gradually reducing the ECM degradation. In this study, we propose a fucoidan-DexMA composite hydrogel, which provides an attractive approach for IVD regeneration.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
- Department of Orthopedics, Lixin County
People’s Hospital, Bozhou, China
| | - Pinghui Zhou
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Bomin Yan
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Meiyao Qi
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Yedan Chen
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
| | - Lijun Shang
- School of Life Sciences, Bengbu Medical
College, Bengbu, China
| | - Jianzhong Guan
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Li Zhang
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Yingji Mao
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
- School of Life Sciences, Bengbu Medical
College, Bengbu, China
| |
Collapse
|
25
|
Zhao X, Ma H, Han H, Zhang L, Tian J, Lei B, Zhang Y. Precision medicine strategies for spinal degenerative diseases: Injectable biomaterials with in situ repair and regeneration. Mater Today Bio 2022; 16:100336. [PMID: 35799898 PMCID: PMC9254127 DOI: 10.1016/j.mtbio.2022.100336] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022]
Abstract
As the population ages, spinal degeneration seriously affects quality of life in middle-aged and elderly patients, and prevention and treatment remain challenging for clinical surgeons. In recent years, biomaterials-based injectable therapeutics have attracted much attention for spinal degeneration treatment due to their minimally invasive features and ability to perform precise repair of irregular defects. However, the precise design and functional control of bioactive injectable biomaterials for efficient spinal degeneration treatment remains a challenge. Although many injectable biomaterials have been reported for the treatment of spinal degeneration, there are few reviews on the advances and effects of injectable biomaterials for spinal degeneration treatment. This work reviews the current status of the design and fabrication of injectable biomaterials, including hydrogels, bone cements and scaffolds, microspheres and nanomaterials, and the current progress in applications for treating spinal degeneration. Additionally, registered clinical trials were also summarized and key challenges and clinical translational prospects for injectable materials for the treatment of spinal degenerative diseases are discussed.
Collapse
|
26
|
Developments on the Smart Hydrogel-Based Drug Delivery System for Oral Tumor Therapy. Gels 2022; 8:gels8110741. [PMID: 36421563 PMCID: PMC9689473 DOI: 10.3390/gels8110741] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
At present, an oral tumor is usually treated by surgery combined with preoperative or postoperative radiotherapies and chemotherapies. However, traditional chemotherapies frequently result in substantial toxic side effects, including bone marrow suppression, malfunction of the liver and kidneys, and neurotoxicity. As a new local drug delivery system, the smart drug delivery system based on hydrogel can control drug release in time and space, and effectively alleviate or avoid these problems. Environmentally responsive hydrogels for smart drug delivery could be triggered by temperature, photoelectricity, enzyme, and pH. An overview of the most recent research on smart hydrogels and their controlled-release drug delivery systems for the treatment of oral cancer is given in this review. It is anticipated that the local drug release method and environment-responsive benefits of smart hydrogels will offer a novel technique for the low-toxicity and highly effective treatment of oral malignancy.
Collapse
|
27
|
Insights into current directions of protein and peptide-based hydrogel drug delivery systems for inflammation. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
28
|
Herrera Quijano MA, Sharma N, Morissette Martin P, Séguin CA, Flynn LE. Development of 2-D and 3-D culture platforms derived from decellularized nucleus pulposus. Front Bioeng Biotechnol 2022; 10:937239. [PMID: 36237211 PMCID: PMC9551564 DOI: 10.3389/fbioe.2022.937239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Bioscaffolds derived from the extracellular matrix (ECM) have shown the capacity to promote regeneration by providing tissue-specific biological instructive cues that can enhance cell survival and direct lineage-specific differentiation. This study focused on the development and characterization of two-dimensional (2-D) and three-dimensional (3-D) cell culture platforms incorporating decellularized nucleus pulposus (DNP). First, a detergent-free protocol was developed for decellularizing bovine nucleus pulposus (NP) tissues that was effective at removing cellular content while preserving key ECM constituents including collagens, glycosaminoglycans, and the cell-adhesive glycoproteins laminin and fibronectin. Next, novel 2-D coatings were generated using the DNP or commercially-sourced bovine collagen type I (COL) as a non-tissue-specific control. In addition, cryo-milled DNP or COL particles were incorporated within methacrylated chondroitin sulphate (MCS) hydrogels as a 3-D cell culture platform for exploring the effects of ECM particle composition. Culture studies showed that the 2-D coatings derived from the DNP could support cell attachment and growth, but did not maintain or rescue the phenotype of primary bovine NP cells, which de-differentiated when serially passaged in monolayer culture. Similarly, while bovine NP cells remained highly viable following encapsulation and 14 days of culture within the hydrogel composites, the incorporation of DNP particles within the MCS hydrogels was insufficient to maintain or rescue changes in NP phenotype associated with extended in vitro culture based on gene expression patterns. Overall, DNP produced with our new decellularization protocol was successfully applied to generate both 2-D and 3-D bioscaffolds; however, further studies are required to assess if these platforms can be combined with additional components of the endogenous NP microenvironment to stimulate regeneration or lineage-specific cell differentiation.
Collapse
Affiliation(s)
- Marco A. Herrera Quijano
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
| | - Nadia Sharma
- Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON, Canada
| | - Pascal Morissette Martin
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Cheryle A. Séguin
- Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- *Correspondence: Lauren E. Flynn, ; Cheryle A. Séguin,
| | - Lauren E. Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON, Canada
- Department of Chemical and Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, ON, Canada
- *Correspondence: Lauren E. Flynn, ; Cheryle A. Séguin,
| |
Collapse
|
29
|
Chang S, Li C, Xu N, Wang J, Jing Z, Cai H, Tian Y, Wang S, Liu Z, Wang X. A sustained release of alendronate from an injectable tetra-PEG hydrogel for efficient bone repair. Front Bioeng Biotechnol 2022; 10:961227. [PMID: 36177182 PMCID: PMC9513246 DOI: 10.3389/fbioe.2022.961227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023] Open
Abstract
Significant efforts on construction of smart drug delivery for developing minimally invasive gelling system to prolong local delivery of bisphosphonates are considered as promising perspectives for the bone-related diseases, which provide the hydrogels with unique bioactivities for bone repair in clinic. Herein, we have constructed an alendronate (ALN)-conjoined injectable tetra-PEG hydrogel with excellent biocompatibility, uniform network, and favorable mechanical properties in one-pot strategy. In views of the quick ammonolysis reaction between N-hydroxysuccinimide (NHS)-ester of tetra-PEG-SG and amine groups of tetra-PEG-NH2 polymer and ALN molecules, the uniform networks were formed within seconds along with the easy injection, favorable biocompatibility and mechanical properties for hydrogel scaffolds. On account of the simultaneous physical encapsulation and chemical linkage of the ALN within the hydrogels, the ALN-conjoined tetra-PEG hydrogel exhibited a sustained drug release delivery that could persistently and effectively facilitate viability, growth, proliferation, and osteogenesis differentiation of stem cells, thereby allowing the consequent adaptation of hydrogels into the bone defects with irregular shapes, which endowed the ALN-conjoined tetra-PEG hydrogel with depot formulation capacity for governing the on-demand release of ALN drugs. Consequently, the findings imply that these drug-based tetra-PEG hydrogels mediate optimal release of therapeutic cargoes and effective promotion of in situ bone regeneration, which will be broadly utilized as therapeutic scaffolds in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Shuai Chang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Chao Li
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Nanfang Xu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Jiedong Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Zehao Jing
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Hong Cai
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Yun Tian
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Shaobo Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Zhongjun Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
- *Correspondence: Zhongjun Liu, ; Xing Wang,
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Zhongjun Liu, ; Xing Wang,
| |
Collapse
|
30
|
Zamboni F, Ren G, Culebras M, O'Driscoll J, O'Dwyer J, Ryan EJ, Collins MN. Curcumin encapsulated polylactic acid nanoparticles embedded in alginate/gelatin bioinks for in situ immunoregulation: Characterization and biological assessment. Int J Biol Macromol 2022; 221:1218-1227. [PMID: 36087752 DOI: 10.1016/j.ijbiomac.2022.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/05/2022]
Abstract
Curcumin is a known naturally occurring anti-inflammatory agent derived from turmeric, and it is commonly used as a herbal food supplement. Here, in order to overcome the inherent hydrophobicity of curcumin (Cur), polylactic acid (PLA) nanoparticles (NPs) were synthesised using a solvent evaporation, and an oil-in-water emulsion method used to encapsulate curcumin. Polymeric NPs also offer the ability to control rate of drug release. The newly synthesised NPs were analysed using a scanning electron microscope (SEM), where results show the NPs range from 50 to 250 nm. NPs containing graded amounts of curcumin (0 %, 0.5 %, and 2 %) were added to cultures of NIH3T3 fibroblast cells for cytotoxicity evaluation using the Alamar Blue assay. Then, the curcumin NPs were incorporated into an alginate/gelatin solution, prior to crosslinking using a calcium chloride solution (200 nM). These hydrogels were then characterised with respect to their chemical, mechanical and rheological properties. Following hydrogel optimization, hydrogels loaded with NP containing 2 % curcumin were selected as a candidate as a bioink for three-dimensional (3D) printing. The biological assessment for these bioinks/hydrogels were conducted using THP-1 cells, a human monocytic cell line. Cell viability and immunomodulation were evaluated using lactate dehydrogenase (LHD) and a tumour necrosis factor alpha (TNF-α) enzyme-linked immunosorbent (ELISA) assay, respectively. Results show that the hydrogels were cytocompatible and supressed the production of TNF-α. These bioactive hydrogels are printable, supress immune cell activation and inflammation showing immense potential for the fabrication of tissue engineering constructs.
Collapse
Affiliation(s)
- Fernanda Zamboni
- Bernal Institute, University of Limerick, Ireland; Health Research Institute, University of Limerick, Ireland
| | - Guang Ren
- Bernal Institute, University of Limerick, Ireland
| | - Mario Culebras
- Institute of Material Science, University of Valencia, Spain
| | | | - Jack O'Dwyer
- School of Engineering, University of Limerick, Ireland
| | - Elizabeth J Ryan
- Health Research Institute, University of Limerick, Ireland; Department of Biological Sciences, University of Limerick, Ireland
| | - Maurice N Collins
- Bernal Institute, University of Limerick, Ireland; Health Research Institute, University of Limerick, Ireland; SFI AMBER, University of Limerick, Ireland.
| |
Collapse
|
31
|
Luneva O, Olekhnovich R, Uspenskaya M. Bilayer Hydrogels for Wound Dressing and Tissue Engineering. Polymers (Basel) 2022; 14:polym14153135. [PMID: 35956650 PMCID: PMC9371176 DOI: 10.3390/polym14153135] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/30/2022] Open
Abstract
A large number of different skin diseases such as hits, acute, and chronic wounds dictate the search for alternative and effective treatment options. The wound healing process requires a complex approach, the key step of which is the choice of a dressing with controlled properties. Hydrogel-based scaffolds can serve as a unique class of wound dressings. Presented on the commercial market, hydrogel wound dressings are not found among proposals for specific cases and have a number of disadvantages—toxicity, allergenicity, and mechanical instability. Bilayer dressings are attracting great attention, which can be combined with multifunctional properties, high criteria for an ideal wound dressing (antimicrobial properties, adhesion and hemostasis, anti-inflammatory and antioxidant effects), drug delivery, self-healing, stimulus manifestation, and conductivity, depending on the preparation and purpose. In addition, advances in stem cell biology and biomaterials have enabled the design of hydrogel materials for skin tissue engineering. To improve the heterogeneity of the cell environment, it is possible to use two-layer functional gradient hydrogels. This review summarizes the methods and application advantages of bilayer dressings in wound treatment and skin tissue regeneration. Bilayered hydrogels based on natural as well as synthetic polymers are presented. The results of the in vitro and in vivo experiments and drug release are also discussed.
Collapse
|
32
|
Jarrah R, Sammak SE, Onyedimma C, Ghaith AK, Moinuddin F, Bhandarkar AR, Siddiqui A, Madigan N, Bydon M. The Role of Alginate Hydrogels as a Potential Treatment Modality for Spinal Cord Injury: A Comprehensive Review of the Literature. Neurospine 2022; 19:272-280. [PMID: 35793929 PMCID: PMC9260541 DOI: 10.14245/ns.2244186.093] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/23/2022] [Indexed: 12/14/2022] Open
Abstract
Objective To comprehensively characterize the utilization of alginate hydrogels as an alternative treatment modality for spinal cord injury (SCI).
Methods An extensive review of the published literature on studies using alginate hydrogels to treat SCI was performed. The review of the literature was performed using electronic databases such as PubMed, EMBASE, and OVID MEDLINE electronic databases. The keywords used were “alginate,” “spinal cord injury,” “biomaterial,” and “hydrogel.”
Results In the literature, we identified a total of 555 rat models that were treated with alginate scaffolds for regenerative biomarkers. Alginate hydrogels were found to be efficient and promising substrates for tissue engineering, drug delivery, neural regeneration, and cellbased therapies for SCI repair. With its ability to act as a pro-regenerative and antidegenerative agent, the alginate hydrogel has the potential to improve clinical outcomes.
Conclusion The emerging developments of alginate hydrogels as treatment modalities may support current and future tissue regenerative strategies for SCI.
Collapse
Affiliation(s)
- Ryan Jarrah
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Sally El Sammak
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Chiduziem Onyedimma
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Abdul Karim Ghaith
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - F.M. Moinuddin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Archis R. Bhandarkar
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Ahad Siddiqui
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Mohamad Bydon
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN, USA
- Corresponding Author Mohamad Bydon Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Zhang W, Chen R, Xu X, Zhu L, Liu Y, Yu X, Tang G. Construction of Biocompatible Hydrogel Scaffolds With a Long-Term Drug Release for Facilitating Cartilage Repair. Front Pharmacol 2022; 13:922032. [PMID: 35784682 PMCID: PMC9245946 DOI: 10.3389/fphar.2022.922032] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/06/2022] [Indexed: 12/24/2022] Open
Abstract
In tissue engineering, hydrogel scaffolds allow various cells to be cultured and grown in vitro and then implanted to repair or replace the damaged areas. Here in this work, kartogenin (KGN), an effectively chondro-inductive non-protein bioactive drug molecule, was incorporated into a composite hydrogel comprising the positively charged chitosan (CS) and methacrylated gelatin (GelMA) polymers to fabricate appropriate microenvironments of bone marrow mesenchymal stem cells (BMSCs) for cartilage regeneration. Based on the combination of physical chain entanglements and chemical crosslinking effects, the resultant GelMA-CS@KGN composite hydrogels possessed favorable network pores and mechanical strength. In vitro cytotoxicity showed the excellent biocompatibility for facilitating the cell growth, adhesion, proliferation, and differentiation. The long-term sustainable KGN release from the hydrogel scaffolds in situ promoted the chondrogenic differentiation that can be employed as an alternative candidate for cartilage tissue regeneration.
Collapse
Affiliation(s)
- Wei Zhang
- Joint Surgery Department, Zhuzhou Central Hospital, Zhuzhou, China
| | - Rui Chen
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiong Xu
- Department of Graduate, Hebei North University, Zhangjiakou, China
| | - Liang Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yanbin Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - XiaoJie Yu
- Department of Orthopedics, Hunan Aerospace Hospital, Changsha, China
- *Correspondence: GuoKe Tang, ; XiaoJie Yu,
| | - GuoKe Tang
- Joint Surgery Department, Zhuzhou Central Hospital, Zhuzhou, China
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: GuoKe Tang, ; XiaoJie Yu,
| |
Collapse
|
34
|
Advances in Hydrogel-Based Microfluidic Blood–Brain-Barrier Models in Oncology Research. Pharmaceutics 2022; 14:pharmaceutics14050993. [PMID: 35631579 PMCID: PMC9144371 DOI: 10.3390/pharmaceutics14050993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 12/10/2022] Open
Abstract
The intrinsic architecture and complexity of the brain restricts the capacity of therapeutic molecules to reach their potential targets, thereby limiting therapeutic possibilities concerning neurological ailments and brain malignancy. As conventional models fail to recapitulate the complexity of the brain, progress in the field of microfluidics has facilitated the development of advanced in vitro platforms that could imitate the in vivo microenvironments and pathological features of the blood–brain barrier (BBB). It is highly desirous that developed in vitro BBB-on-chip models serve as a platform to investigate cancer metastasis of the brain along with the possibility of efficiently screening chemotherapeutic agents against brain malignancies. In order to improve the proficiency of BBB-on-chip models, hydrogels have been widely explored due to their unique physical and chemical properties, which mimic the three-dimensional (3D) micro architecture of tissues. Hydrogel-based BBB-on-chip models serves as a stage which is conducive for cell growth and allows the exchange of gases and nutrients and the removal of metabolic wastes between cells and the cell/extra cellular matrix (ECM) interface. Here, we present recent advancements in BBB-on-chip models targeting brain malignancies and examine the utility of hydrogel-based BBB models that could further strengthen the future application of microfluidic devices in oncology research.
Collapse
|
35
|
Quantitative Photoacoustic Reconstruction of the Optical Properties of Intervertebral Discs Using a Gradient Descent Scheme. PHOTONICS 2022. [DOI: 10.3390/photonics9020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The intervertebral discs (IVD) are among the essential organs of the human body, ensuring the mobility of the spine. These organs possess a high proportion of water. However, as the discs age, this content decreases, which can potentially lead to various diseases called degenerative disc diseases. This water content is therefore an important indicator of the well-being of the disc. In this paper, we propose photoacoustic imaging as a means of probing a disc and quantitatively recovering its molecular composition, which should allow concluding on its state. An adjoint-assisted gradient descent scheme is implemented to recover the optical absorption coefficient in the disc, from which, if spectroscopic measurements are performed, the molecular composition can be deduced. The algorithm was tested on synthetic measurements. A realistic numerical phantom was built from magnetic resonance imaging of an actual IVD of a pig. A simplified experiment, with a single laser source, was performed. Results show the feasibility of using photoacoustics imaging to probe IVDs. The influences of exact and approximate formulations of the gradient are studied. The impact of noise on the reconstructions is also evaluated.
Collapse
|
36
|
Øvrebø Ø, Perale G, Wojciechowski JP, Echalier C, Jeffers JRT, Stevens MM, Haugen HJ, Rossi F. Design and clinical application of injectable hydrogels for musculoskeletal therapy. Bioeng Transl Med 2022; 7:e10295. [PMID: 35600661 PMCID: PMC9115710 DOI: 10.1002/btm2.10295] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/25/2022] Open
Abstract
Musculoskeletal defects are an enormous healthcare burden and source of pain and disability for individuals. With an aging population, the proportion of individuals living with these medical indications will increase. Simultaneously, there is pressure on healthcare providers to source efficient solutions, which are cheaper and less invasive than conventional technology. This has led to an increased research focus on hydrogels as highly biocompatible biomaterials that can be delivered through minimally invasive procedures. This review will discuss how hydrogels can be designed for clinical translation, particularly in the context of the new European Medical Device Regulation (MDR). We will then do a deep dive into the clinically used hydrogel solutions that have been commercially approved or have undergone clinical trials in Europe or the United States. We will discuss the therapeutic mechanism and limitations of these products. Due to the vast application areas of hydrogels, this work focuses only on treatments of cartilage, bone, and the nucleus pulposus. Lastly, the main steps toward clinical translation of hydrogels as medical devices are outlined. We suggest a framework for how academics can assist small and medium MedTech enterprises conducting the initial clinical investigation and post‐market clinical follow‐up required in the MDR. It is evident that the successful translation of hydrogels is governed by acquiring high‐quality pre‐clinical and clinical data confirming the device mechanism of action and safety.
Collapse
Affiliation(s)
- Øystein Øvrebø
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilanoItaly
- Department of BiomaterialsInstitute of Clinical Dentistry, University of OsloOsloNorway
- Material Biomimetic ASOslo Science ParkOsloNorway
| | - Giuseppe Perale
- Industrie Biomediche Insubri SAMezzovico‐ViraSwitzerland
- Faculty of Biomedical SciencesUniversity of Southern SwitzerlandLuganoSwitzerland
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyViennaAustria
| | - Jonathan P. Wojciechowski
- Department of MaterialsImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
- Institute of Biomedical EngineeringImperial College LondonLondonUK
| | - Cécile Echalier
- Department of MaterialsImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
- Institute of Biomedical EngineeringImperial College LondonLondonUK
- Hybrid Technology Hub, Centre of ExcellenceInstitute of Basic Medical Science, University of OsloOsloNorway
| | | | - Molly M. Stevens
- Department of MaterialsImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
- Institute of Biomedical EngineeringImperial College LondonLondonUK
| | - Håvard J. Haugen
- Department of BiomaterialsInstitute of Clinical Dentistry, University of OsloOsloNorway
- Material Biomimetic ASOslo Science ParkOsloNorway
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilanoItaly
| |
Collapse
|
37
|
Yu L, Liu Y, Wu J, Wang S, Yu J, Wang W, Ye X. Genipin Cross-Linked Decellularized Nucleus Pulposus Hydrogel-Like Cell Delivery System Induces Differentiation of ADSCs and Retards Intervertebral Disc Degeneration. Front Bioeng Biotechnol 2022; 9:807883. [PMID: 35004657 PMCID: PMC8733700 DOI: 10.3389/fbioe.2021.807883] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/03/2021] [Indexed: 01/08/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is the pathological basis of disc degenerative diseases (DDD). Reduction in the number of cells and degeneration of the extracellular matrix (ECM) in the nucleus pulposus (NP) are characteristics of IDD. Bio-hydrogel combined with stem cell transplantation is a promising treatment. Injectable ECM hydrogels have good biological activity and in-situ gelatinization. However, its biomechanics and stability are insufficient to provide adequate mechanical support for intervertebral discs and to maintain the long-term differential stimulus for seeded stem cells. In our study, we developed genipin cross-linked decellularized nucleus pulposus hydrogel (GDH) as delivery system. We evaluated the mechanical properties, stability, biocompatibility, and differentiation induction of GDH cross-linked with different concentrations of genipin in vitro. The GDH-loaded adipose-derived mesenchymal stem cells (ADSCs) (GDHA) were injected into the rat degenerated coccygeal intervertebral disc. The effect of intervertebral disc regeneration in vivo was evaluated. The results showed that GDH with 0.02% of genipin had similar elastic modulus to human nucleus pulposus, good biocompatibility, and inducibility of expressing NP-related genes. In vivo studies showed that GDHA improved the survival of ADSCs and improved the intervertebral height, MRI index, and histological grading score. In conclusion, GDH, as an outstanding bio-hydrogel cell delivery system, has the therapeutic potential for retarding IDD.
Collapse
Affiliation(s)
- Lei Yu
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yi Liu
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianxin Wu
- Department of Orthopedics, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shuang Wang
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiangming Yu
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Weiheng Wang
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaojian Ye
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
38
|
Hossain L, Raghuwanshi VS, Tanner J, Garnier G. Modulating nanocellulose hydrogels and cryogels strength by crosslinking and blending. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
39
|
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: 3.0] [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
|
40
|
Naranda J, Bračič M, Vogrin M, Maver U. Recent Advancements in 3D Printing of Polysaccharide Hydrogels in Cartilage Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3977. [PMID: 34300896 PMCID: PMC8305911 DOI: 10.3390/ma14143977] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022]
Abstract
The application of hydrogels coupled with 3-dimensional (3D) printing technologies represents a modern concept in scaffold development in cartilage tissue engineering (CTE). Hydrogels based on natural biomaterials are extensively used for this purpose. This is mainly due to their excellent biocompatibility, inherent bioactivity, and special microstructure that supports tissue regeneration. The use of natural biomaterials, especially polysaccharides and proteins, represents an attractive strategy towards scaffold formation as they mimic the structure of extracellular matrix (ECM) and guide cell growth, proliferation, and phenotype preservation. Polysaccharide-based hydrogels, such as alginate, agarose, chitosan, cellulose, hyaluronan, and dextran, are distinctive scaffold materials with advantageous properties, low cytotoxicity, and tunable functionality. These superior properties can be further complemented with various proteins (e.g., collagen, gelatin, fibroin), forming novel base formulations termed "proteo-saccharides" to improve the scaffold's physiological signaling and mechanical strength. This review highlights the significance of 3D bioprinted scaffolds of natural-based hydrogels used in CTE. Further, the printability and bioink formation of the proteo-saccharides-based hydrogels have also been discussed, including the possible clinical translation of such materials.
Collapse
Affiliation(s)
- Jakob Naranda
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia;
| | - Matej Bračič
- Faculty of Mechanical Engineering, University of Maribor, SI-2000 Maribor, Slovenia;
| | - Matjaž Vogrin
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia;
- Department of Orthopaedics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
- Department of Pharmacology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
| |
Collapse
|
41
|
Carvalho SG, Silvestre ALP, Martins Dos Santos A, Fonseca-Santos B, Rodrigues WD, Palmira Daflon Gremião M, Chorilli M, Villanova JCO. Polymeric-based drug delivery systems for veterinary use: State of the art. Int J Pharm 2021; 604:120756. [PMID: 34058307 DOI: 10.1016/j.ijpharm.2021.120756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/11/2021] [Accepted: 05/26/2021] [Indexed: 01/03/2023]
Abstract
One of the challenges to the success of veterinary pharmacotherapy is the limited number of drugs and dosage forms available exclusively to this market, due to the interspecies variability of animals, such as anatomy, physiology, pharmacokinetics, and pharmacodynamics. For this reason, studies in this area have become a highlight, since they are still scarce in comparison with those on human drug use. To overcome many limitations related to the bioavailability, efficacy, and safety of pharmacotherapy in animals, especially livestock and domestic animals, polymers-based drug delivery systems are promising tools if they guarantee greater selectivity and less toxicity in dosage forms. In addition, these tools may be developed according to the great interspecies variability. To contribute to these discussions, this paper provides an updated review of the major polymer-based drug delivery systems projected for veterinary use. Traditional and innovative drug delivery systems based on polymers are presented, with an emphasis on films, microparticles, micelles, nanogels, nanoparticles, tablets, implants and hydrogel-based drug delivery systems. We discuss important concepts for the veterinarian about the mechanisms of drug release and, for the pharmacist, the advantages in the development of pharmaceutical forms for the animal population. Finally, challenges and opportunities are presented in the field of pharmaceutical dosage forms for veterinary use in response to the interests of the pharmaceutical industry.
Collapse
Affiliation(s)
- Suzana Gonçalves Carvalho
- School of Pharmaceutical Sciences, São Paulo State University, UNESP, Department of Drugs and Medicines, 14800-903 Araraquara, SP, Brazil.
| | - Amanda Letícia Polli Silvestre
- School of Pharmaceutical Sciences, São Paulo State University, UNESP, Department of Drugs and Medicines, 14800-903 Araraquara, SP, Brazil
| | - Aline Martins Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University, UNESP, Department of Drugs and Medicines, 14800-903 Araraquara, SP, Brazil
| | - Bruno Fonseca-Santos
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), 13083-871 Campinas, SP, Brazil
| | - Winner Duque Rodrigues
- School of Pharmaceutical Sciences, São Paulo State University, UNESP, Department of Drugs and Medicines, 14800-903 Araraquara, SP, Brazil
| | - Maria Palmira Daflon Gremião
- School of Pharmaceutical Sciences, São Paulo State University, UNESP, Department of Drugs and Medicines, 14800-903 Araraquara, SP, Brazil.
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University, UNESP, Department of Drugs and Medicines, 14800-903 Araraquara, SP, Brazil.
| | - Janaína Cecília Oliveira Villanova
- Laboratory of Pharmaceutical Production, Department of Pharmacy and Nutrition, Federal University of Espirito Santo (UFES), 29500-000 Alegre, ES, Brazil.
| |
Collapse
|
42
|
Ashinsky B, Smith HE, Mauck RL, Gullbrand SE. Intervertebral disc degeneration and regeneration: a motion segment perspective. Eur Cell Mater 2021; 41:370-380. [PMID: 33763848 PMCID: PMC8607668 DOI: 10.22203/ecm.v041a24] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Back and neck pain have become primary reasons for disability and healthcare spending globally. While the causes of back pain are multifactorial, intervertebral disc degeneration is frequently cited as a primary source of pain. The annulus fibrosus (AF) and nucleus pulposus (NP) subcomponents of the disc are common targets for regenerative therapeutics. However, disc degeneration is also associated with degenerative changes to adjacent spinal tissues, and successful regenerative therapies will likely need to consider and address the pathology of adjacent spinal structures beyond solely the disc subcomponents. This review summarises the current state of knowledge in the field regarding associations between back pain, disc degeneration, and degeneration of the cartilaginous and bony endplates, the AF-vertebral body interface, the facet joints and spinal muscles, in addition to a discussion of regenerative strategies for treating pain and degeneration from a whole motion segment perspective.
Collapse
Affiliation(s)
| | | | | | - S E Gullbrand
- Corporal Michael J. Crescenz VA Medical Centre, Research, Building 21, Rm A214, 3900 Woodland Ave, Philadelphia, PA 19104,
| |
Collapse
|
43
|
Wang J, Wang L, Gao Y, Zhang Z, Huang X, Han T, Liu B, Zhang Y, Li Y, Zhang L. Synergistic Therapy of Celecoxib-Loaded Magnetism-Responsive Hydrogel for Tendon Tissue Injuries. Front Bioeng Biotechnol 2020; 8:592068. [PMID: 33330423 PMCID: PMC7729092 DOI: 10.3389/fbioe.2020.592068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/12/2020] [Indexed: 11/13/2022] Open
Abstract
Tendon tissue injury is very common and always associated with pain, tissue swelling and even malformation if not treated on time. Traditional therapeutic strategies, such as cryotherapy, electrical therapy, ultrasound therapy and anti-inflammatory drug, are still unsatisfying. In this work, a synergistic therapy, based on the combination of celecoxib drug and pulsed electromagnetic field (PEMF) regimens, was developed for the treatment of tendon injury. This celecoxib-loaded magnetism-responsive hydrogel dressing (gelatin/Fe3O4/celecoxib) showed good biocompatibility and coordinated drug release behavior under the PEMF, which could effectively reduce the inflammatory reaction of macrophage cells with the incremental proportion of M2 macrophages at the injury site. CatWalk gait analysis further verified this synergistic effect of combination therapy for achieving the outstanding recovery of the injured tendon tissue. Thus, this magnetism-responsive hydrogel may represent a promising alternative strategy in clinics for promoting tendon healing.
Collapse
Affiliation(s)
- Jingxin Wang
- Department of Rehabilitation, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Likang Wang
- Department of Rehabilitation Medicine, The Third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yueming Gao
- Department of Rehabilitation Medicine, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhao Zhang
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Xiaofeng Huang
- Department of Endocrinology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Tong Han
- Department of Rehabilitation, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Biyuan Liu
- Department of Rehabilitation, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yujie Zhang
- Department of Epidemiology, School of Public Health Southern Medical University, Guangzhou, China
| | - Yilan Li
- Department of Rehabilitation, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lining Zhang
- Department of Rehabilitation Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
44
|
Zhang M, Liang J, Yang Y, Liang H, Jia H, Li D. Current Trends of Targeted Drug Delivery for Oral Cancer Therapy. Front Bioeng Biotechnol 2020; 8:618931. [PMID: 33425881 PMCID: PMC7793972 DOI: 10.3389/fbioe.2020.618931] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/19/2020] [Indexed: 12/29/2022] Open
Abstract
Oral cancer is an aggressive tumor that invades the local tissue and can cause metastasis and high mortality. Conventional treatment strategies, e.g., surgery, chemotherapy, and radiation therapy alone or in combinations, possess innegligible issues, and significant side and adverse effects for the clinical applications. Currently, targeting drug delivery is emerging as an effective approach for oral delivery of different therapeutics. Herein we provide a state-of-the-art review on the current progress of targeting drug delivery for oral cancer therapy. Variously oral delivery systems including polymeric/inorganic nanoparticles, liposomes, cyclodextrins, nanolipids, and hydrogels-based forms are emphasized and discussed, and biomimetic systems with respect to oral delivery like therapeutic vitamin, exosomes, proteins, and virus-like particles are also described with emphasis on the cancer treatment. A future perspective is also provided to highlight the existing challenges and possible resolution toward clinical translation of current oral cancer therapies.
Collapse
Affiliation(s)
- Mingming Zhang
- Strategic Support Force Characteristic Medical Center of Chinese People's Liberation Army, Beijing, China
| | - Jianqin Liang
- The 8th Medical Center, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Yanyu Yang
- College of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
| | - Huize Liang
- Strategic Support Force Characteristic Medical Center of Chinese People's Liberation Army, Beijing, China
| | - Huaping Jia
- Strategic Support Force Characteristic Medical Center of Chinese People's Liberation Army, Beijing, China
| | - Dawei Li
- The 4th Medical Center, General Hospital of the Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
45
|
Li D, Chen J, Wang X, Zhang M, Li C, Zhou J. Recent Advances on Synthetic and Polysaccharide Adhesives for Biological Hemostatic Applications. Front Bioeng Biotechnol 2020; 8:926. [PMID: 32923431 PMCID: PMC7456874 DOI: 10.3389/fbioe.2020.00926] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Rapid hemostasis and formation of stable blood clots are very important to prevent massive blood loss from the excessive bleeding for living body, but their own clotting process cannot be completed in time for effective hemostasis without the help of hemostatic materials. In general, traditionally suturing and stapling techniques for wound closure are prone to cause the additional damages to the tissues, activated inflammatory responses, short usage periods and inevitable second operations in clinical applications. Especially for the large wounds that require the urgent closure of fluids or gases, these conventional closure methods are far from enough. To address these problems, various tissue adhesives, sealants and hemostatic materials are placed great expectation. In this review, we focused on the development of two main categories of tissue adhesive materials: synthetic polymeric adhesives and naturally derived polysaccharide adhesives. Research of the high performance of hemostatic adhesives with strong adhesion, better biocompatibility, easy usability and cheap price is highly demanded for both scientists and clinicians, and this review is also intended to provide a comprehensive summarization and inspiration for pursuit of more advanced hemostatic adhesives for biological fields.
Collapse
Affiliation(s)
- Dawei Li
- Eighth Medical Center of the General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Jing Chen
- Department of Orthopedics, Aerospace Center Hospital, Beijing, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mingming Zhang
- The People’s Liberation Army Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Chunlin Li
- Eighth Medical Center of the General Hospital of the Chinese People’s Liberation Army, Beijing, China
| | - Jin Zhou
- Eighth Medical Center of the General Hospital of the Chinese People’s Liberation Army, Beijing, China
| |
Collapse
|