1
|
Yang S, Soheilmoghaddam F, Pivonka P, Li J, Rudd S, Yeo T, Tu J, Zhu Y, Cooper-White JJ. Engineering Intervertebral Disc Regeneration: Biomaterials, Cell Sources and Animal Models. Cell Prolif 2025:e70046. [PMID: 40389238 DOI: 10.1111/cpr.70046] [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: 03/01/2025] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 05/21/2025] Open
Abstract
Intervertebral disc (IVD) degeneration is an age-related problem triggering chronic spinal issues, such as low back pain and IVD herniation. Standard surgical treatment for such spinal issues is the removal of the degenerated or herniated IVD and fusion of adjacent vertebrae to stabilise the joint and locally decompress the spinal cord and/or nerve roots to relieve pain. However, a key challenge of current surgical strategies is the increasing risk of adjacent segment degeneration due to the disruption of native biomechanics of the functional spinal unit, dominated by the loss of the IVD. In the past two decades, research has focused on developing a number of bioengineering approaches to repair and regenerate the IVD; in particular, tissue engineering of the IVD, using bioscaffolds and stem cells represents a promising area. This review highlights the current tissue engineering approaches utilising biomaterials, animal models and cell sources for IVD regeneration and discusses future opportunities.
Collapse
Affiliation(s)
- Sidong Yang
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Hebei International Joint Research Centre for Spine Diseases, Shijiazhuang, China
| | - Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
| | - Peter Pivonka
- School of Mechanical Medical & Process Engineering, Queensland University of Technology, Brisbane City, Queensland, Australia
| | - Joan Li
- Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Samuel Rudd
- School of Chemical Engineering, The University of Queensland, St Lucia, Queensland, Australia
| | - Trifanny Yeo
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- Institute for Health Innovation & Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Ji Tu
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Yibo Zhu
- School of Chemical Engineering, The University of Queensland, St Lucia, Queensland, Australia
| | - Justin J Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, Queensland, Australia
- School of Chemical Engineering, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
2
|
Keshavarz S, Alavi CE, Aghayan H, Jafari-Shakib R, Vojoudi E. Advancements in Degenerative Disc Disease Treatment: A Regenerative Medicine Approach. Stem Cell Rev Rep 2025:10.1007/s12015-025-10882-z. [PMID: 40232618 DOI: 10.1007/s12015-025-10882-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Regenerative medicine represents a transformative approach to treating nucleus pulposus degeneration and offers hope for patients suffering from chronic low back pain due to disc degeneration. By focusing on restoring the natural structure and function of the nucleus pulposus rather than merely alleviating symptoms, these innovative therapies hold the potential to significantly improve patient outcomes. As research continues to advance in this field, we may soon witness a paradigm shift in how we approach spinal health and degenerative disc disease. The main purpose of this review is to provide an overview of the various regenerative approaches that target the restoration of the nucleus pulposus, a primary site for initiation of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Samaneh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Cyrus Emir Alavi
- Department of Anesthesiology, Neuroscience Research Center, Avicenna University Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamidreza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari-Shakib
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, P.O.Box 41635 - 3363, Rasht, Iran.
| | - Elham Vojoudi
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
3
|
Carvalho AF, Pereira T, Oliveira C, Figueiredo P, Carvalho A, Pereira DM, Hilliou L, Bañobre-López M, Xu B, Ferreira PMT, Martins JA. Tripeptides Featuring Dehydrophenylalanine and Homophenylalanine: Homo- Versus Hetero-Chirality and Sequence Effects on Self-Assembly and Gelation. Gels 2025; 11:164. [PMID: 40136869 PMCID: PMC11942182 DOI: 10.3390/gels11030164] [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: 01/12/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Over the years, our research group developed dehydrodipeptides N-capped with aromatic moieties as protease-resistant efficacious hydrogelators, affording self-assembled hydrogels at low (critical) concentrations. Dehydrotripeptides, with different dipeptide sequences and (D,L) stereochemistry, open a wider chemical space for the development of self-assembled soft nanomaterials. In this work, a small library of N-succinylated dehydrotripeptides containing a C-terminal dehydrophenylalanine (∆Phe) residue and a scrambled dipeptide sequence with phenylalanine (Phe) and homophenylalanine (Hph) (L-Phe-L,D-Hph and L,D-Hph-L-Phe) was synthesized and characterized as a potential hydrogelator. Two pairs of diastereomeric tripeptides were synthesized, both as C-protected methyl esters and as deprotected dicarboxylic acids. Peptides with the sequence Hph-Phe-ΔPhe were obtained as a pair (D,L,Z)/(L,L,Z) of diastereomers. Their scrambled sequence analogues Phe-Hph-ΔPhe were obtained also as a diastereomeric (L,D,Z)/(L,L,Z) pair. The effect of stereochemistry (homo- vs. hetero-chirality) and sequence (Phe-∆Phe vs. Hph-∆Phe motif) on the self-assembly, biocompatibility, gelation and rheological properties of the hydrogels was studied in this work. Accessible, both as C-protected methyl esters and as dicarboxylic acids, N-succinylated dehydrotripeptides are interesting molecular architectures for the development of supramolecular nanomaterials. Interestingly, our results do not comply with the well-documented proposition that heterochiral peptides display much higher self-assembly propensity and gelation ability than their homochiral counterparts. Further studies will be necessary to fully understand the interplay between peptide sequence and homo- and hetero-chirality on peptide self-assembly and on the properties of their supramolecular materials.
Collapse
Affiliation(s)
- André F. Carvalho
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| | - Teresa Pereira
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| | - Carlos Oliveira
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| | - Pedro Figueiredo
- CNC—Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Alexandra Carvalho
- CNC—Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- Almac Sciences, Department of Biocatalysis and Isotope Chemistry, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, UK
| | - David M. Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n 228, 4050-313 Porto, Portugal
| | - Loic Hilliou
- Institute for Polymers and Composites, University of Minho, 4800-058 Guimarães, Portugal;
| | - Manuel Bañobre-López
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal;
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | | | - José A. Martins
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| |
Collapse
|
4
|
Soliman MAN, Khedr A, Sahota T, Armitage R, Allan R, Laird K, Allcock N, Ghuloum FI, Amer MH, Alazragi R, Edwards‐Gayle CJC, Wychowaniec JK, Vargiu AV, Elsawy MA. Unraveling the Atomistic Mechanism of Electrostatic Lateral Association of Peptide β-Sheet Structures and Its Role in Nanofiber Growth and Hydrogelation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408213. [PMID: 39780584 PMCID: PMC11817957 DOI: 10.1002/smll.202408213] [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: 09/10/2024] [Revised: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Guiding molecular assembly of peptides into rationally engineered nanostructures remains a major hurdle against the development of functional peptide-based nanomaterials. Various non-covalent interactions come into play to drive the formation and stabilization of these assemblies, of which electrostatic interactions are key. Here, the atomistic mechanisms by which electrostatic interactions contribute toward controlling self-assembly and lateral association of ultrashort β-sheet forming peptides are deciphered. Our results show that this is governed by charge distribution and ionic complementarity, both affecting the interaction patterns between charged residues: terminal, core, and/or terminal-to-core attraction/repulsion. Controlling electrostatic interactions enabled fine-tuning nanofiber morphology for the 16 examined peptides, resulting into versatile nanostructures ranging from extended thin fibrils and thick bundles to twisted helical "braids" and short pseudocrystalline nanosheets. This in turn affected the physical appearance and viscoelasticity of the formed materials, varying from turbid colloidal dispersions and viscous solutions to soft and stiff self-supportive hydrogels, as revealed from oscillatory rheology. Atomistic mechanisms of electrostatic interaction patterns were confirmed by molecular dynamic simulations, validating molecular and nanoscopic characterization of the developed materials. In essence, detailed mechanisms of electrostatic interactions emphasizing the impact of charge distribution and ionic complementarity on self-assembly, nanostructure formation, and hydrogelation are reported.
Collapse
Affiliation(s)
- Mohamed A. N. Soliman
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
- Department of Pharmaceutics and Industrial PharmacyFaculty of PharmacyCairo UniversityCairo11562Egypt
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
- Department of Pharmaceutics and Industrial PharmacyFaculty of PharmacyZagazig UniversityZagazigEgypt
| | - Tarsem Sahota
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
| | - Rachel Armitage
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
- School of Archaeology and Ancient HistoryUniversity of LeicesterLeicesterLE1 7RHUK
| | - Raymond Allan
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
| | - Katie Laird
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
| | - Natalie Allcock
- Electron Microscopy Facility Core Biotechnology ServicesCollege of Life SciencesUniversity of LeicesterLeicesterLE1 7RHUK
| | - Fatmah I. Ghuloum
- Division of Cell Matrix and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterOxford RoadManchesterM13 9PLUK
| | - Mahetab H. Amer
- Division of Cell Matrix and Regenerative MedicineSchool of Biological SciencesUniversity of ManchesterOxford RoadManchesterM13 9PLUK
| | - Reem Alazragi
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
- Department of Biological ScienceCollege of ScienceUniversity of JeddahJeddah21493Saudi Arabia
| | | | | | - Attilio V. Vargiu
- Physics DepartmentUniversity of Cagliaris.p. 8km. 0.700Monserrato09042Italy
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical InnovationLeicester School of PharmacyDe Montfort UniversityThe GatewayLeicesterLE1 9BHUK
- Division of Pharmacy and OptometrySchool of Health SciencesUniversity of ManchesterOxford RoadManchesterM13 9PLUK
| |
Collapse
|
5
|
Guo C, Jiao X, Du X, Zhang T, Peng B, Xu B. Application of Self-Healing Hydrogels in the Treatment of Intervertebral Disc Degeneration. J Biomed Mater Res B Appl Biomater 2025; 113:e35532. [PMID: 39842850 DOI: 10.1002/jbm.b.35532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/25/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
Intervertebral disc degeneration (IDD) is one of the leading causes of chronic pain and disability, and traditional treatment methods often struggle to restore its complex biomechanical properties. This article explores the innovative application of self-healing hydrogels in the treatment of IDD, offering new hope for disc repair due to their exceptional self-repair capabilities and adaptability. As a key support structure in the human body, intervertebral discs are often damaged by trauma or degenerative changes. Self-healing hydrogels not only mimic the mechanical properties of natural intervertebral discs but also self-repair when damaged, thereby maintaining stable functionality. This article reviews the self-healing mechanisms and design strategies of self-healing hydrogels and, for the first time, outlines their potential in the treatment of IDD. Furthermore, the article looks forward to future developments in the field, including intelligent material design, multifunctional integration, encapsulation and release of bioactive molecules, and innovative combinations with tissue engineering and stem cell therapy, offering new perspectives and strategies for IDD treatment.
Collapse
Affiliation(s)
- Cunliang Guo
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinyi Jiao
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoxun Du
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | | - Bing Peng
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | |
Collapse
|
6
|
Sanjaykumar SG, Malviya R, Srivastava S, Ahmad I, Uniyal P, Singh B, Nisar N. Chitosan-Peptide Composites for Tissue Engineering Applications: Advances in Treatment Strategies. Curr Protein Pept Sci 2025; 26:185-200. [PMID: 39350425 DOI: 10.2174/0113892037323136240910052119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 04/09/2025]
Abstract
One of the most well-known instances of an interdisciplinary subject is tissue engineering, where experts from many backgrounds collaborate to address important health issues and improve people's quality of life. Many researchers are interested in using chitosan and its derivatives as an alternative to fabricating scaffold engineering and skin grafts in tissue because of its natural abundance, affordability, biodegradability, biocompatibility, and wound healing properties. Nanomaterials based on peptides can provide cells with the essential biological cues required to promote cellular adhesion and are easily fabricated. Due to such worthy properties of chitosan and peptide, they find their application in tissue engineering and regeneration processes. The implementation of hybrids of chitosan and peptide is increasing in the field of tissue engineering and scaffolding for improved cellular adherence and bioactivity. This review covers the individual applications of peptide and chitosan in tissue engineering and further discusses the role of their conjugates in the same. Here, the recent findings are also discussed, along with studies involving the use of these hybrids in tissue engineering applications.
Collapse
Affiliation(s)
- Swati Gupta Sanjaykumar
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Bhupinder Singh
- Department of Law, Sharda University, Greater Noida, U.P., India
| | - Nazima Nisar
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
7
|
Xin Y, Ligorio C, O'brien M, Collins R, Dong S, Miller AF, Saiani A, Gough JE. Effect of supramolecular peptide hydrogel scaffold charge on HepG2 viability and spheroid formation. J Mater Chem B 2024; 12:12553-12566. [PMID: 39502032 DOI: 10.1039/d4tb01701c] [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: 12/12/2024]
Abstract
Supramolecular bioinspired self-assembling peptide hydrogel (SAPH) scaffolds represent a class of fully defined synthetic materials whose chemical and mechanical properties can be finely engineered. In this study, the relationship between SAPHs physicochemical properties and HepG2 cells viability, spheroid formation and function are discussed. We first report that negatively charged SAPHs promote hepatocyte proliferation and spheroids formation in vitro 3D culture while positively charged SAPHs lead to hepatocyte death irrespective of the hydrogel mechanical properties. More specifically HepG2 cultured in 3D in E(FKFE)2 negatively charged SAPH maintained a differentiated phenotype and assembled into well-defined spheroids with strong cell-cell interactions. Furthermore, HepG2 spheroids responded to acetaminophen exposure with upregulation of key CYP450 enzymes expression clearly showing their potential for drug toxicity testing. These findings demonstrate how fine-tuned functional SAPH scaffolds can be used to identify key scaffolds parameters affecting cells. In this case we demonstrated the potential of negatively charged SAPHs for the 3D culture of HepG2 with potential applications in drug screening.
Collapse
Affiliation(s)
- Yu Xin
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| | - Cosimo Ligorio
- Department of Materials & Manchester Institute of Biotechnology, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Marie O'brien
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| | - Richard Collins
- Electron Microscopy Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Siyuan Dong
- Department of Chemical Engineering & Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester, UK
| | - Aline F Miller
- Department of Chemical Engineering & Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester, UK
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Julie E Gough
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| |
Collapse
|
8
|
Chen X, Jing S, Xue C, Guan X. Progress in the Application of Hydrogels in Intervertebral Disc Repair: A Comprehensive Review. Curr Pain Headache Rep 2024; 28:1333-1348. [PMID: 38985414 PMCID: PMC11666692 DOI: 10.1007/s11916-024-01296-6] [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] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE OF REVIEW Intervertebral disc degeneration (IVDD) is a common orthopaedic disease and an important cause of lower back pain, which seriously affects the work and life of patients and causes a large economic burden to society. The traditional treatment of IVDD mainly involves early pain relief and late surgical intervention, but it cannot reverse the pathological course of IVDD. Current studies suggest that IVDD is related to the imbalance between the anabolic and catabolic functions of the extracellular matrix (ECM). Anti-inflammatory drugs, bioactive substances, and stem cells have all been shown to improve ECM, but traditional injection methods face short half-life and leakage problems. RECENT FINDINGS The good biocompatibility and slow-release function of polymer hydrogels are being noticed and explored to combine with drugs or bioactive substances to treat IVDD. This paper introduces the pathophysiological mechanism of IVDD, and discusses the advantages, disadvantages and development prospects of hydrogels for the treatment of IVDD, so as to provide guidance for future breakthroughs in the treatment of IVDD.
Collapse
Affiliation(s)
- Xin Chen
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Shaoze Jing
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Chenhui Xue
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xiaoming Guan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| |
Collapse
|
9
|
Warren JP, Coe RH, Culbert MP, Dixon AR, Miles DE, Mengoni M, Beales PA, Wilcox RK. Injectable peptide-glycosaminoglycan hydrogels for soft tissue repair: in vitro assessment for nucleus augmentation. MATERIALS ADVANCES 2024; 5:8665-8672. [PMID: 39421698 PMCID: PMC11474259 DOI: 10.1039/d4ma00613e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024]
Abstract
We report the development of peptide-glycosaminoglycan hydrogels as injectable biomaterials for load-bearing soft tissue repair. The hydrogels are injectable as a liquid for clinical delivery, rapidly form a gel in situ, and mimic the osmotic swelling behaviour of natural tissue. We used a new in vitro model to demonstrate their application as a nucleus augmentation material for the treatment of intervertebral disc degeneration. Our study compared a complex lab gel preparation method to a simple clinical benchtop process. We showed pH differences did not significantly affect gel formation, and temperature variations had no impact on gel performance. Rheological results demonstrated consistency after benchtop mixing or needle injection. In our in vitro disc degeneration model, we established that peptide augmentation could restore the native biomechanical properties. This suggests the feasibility of minimally invasive peptide-GAG gel delivery, maintaining consistent properties across temperature and needle sizes while restoring disc height and stiffness in vitro.
Collapse
Affiliation(s)
- James P Warren
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
- School of Chemistry, University of Leeds Leeds LS2 9JT UK
| | - Ruth H Coe
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
| | - Matthew P Culbert
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
- School of Chemistry, University of Leeds Leeds LS2 9JT UK
| | - Andrew R Dixon
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
| | - Danielle E Miles
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
- School of Chemistry, University of Leeds Leeds LS2 9JT UK
| | - Marlène Mengoni
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
| | - Paul A Beales
- School of Chemistry, University of Leeds Leeds LS2 9JT UK
| | - Ruth K Wilcox
- Institute of Medical and Biological Engineering, School of Mechanical Engineering UK
| |
Collapse
|
10
|
Wang J, Zhang Y, Huang Y, Hao Z, Shi G, Guo L, Chang C, Li J. Application trends and strategies of hydrogel delivery systems in intervertebral disc degeneration: A bibliometric review. Mater Today Bio 2024; 28:101251. [PMID: 39318370 PMCID: PMC11421353 DOI: 10.1016/j.mtbio.2024.101251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/16/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Hydrogels are widely used to explore emerging minimally invasive strategies for intervertebral disc degeneration (IVDD) due to their suitability as drug and cell delivery vehicles. There has been no review of the latest research trends and strategies of hydrogel delivery systems in IVDD for the last decade. In this study, we identify the application trends and strategies in this field through bibliometric analysis, including aspects such as publication years, countries and institutions, authors and publications, and co-occurrence of keywords. The results reveal that the literature in this field has been receiving increasing attention with a trend of growth annually. Subsequently, the hotspots of hydrogels in this field were described and discussed in detail, and we proposed the "four core factors", hydrogels, cells, cell stimulators, and microenvironmental regulation, required for a multifunctional hydrogel for IVDD. Finally, we discuss the popular and emerging mechanistic strategies of hydrogel therapy for IVDD in terms of five aspects: fundamental pathologic changes in IVDD, counteracting cellular senescence, counteracting cell death, improving organelle function, and replenishing exogenous cells. This study provides a reference and a new perspective for future research in this urgently needed field.
Collapse
Affiliation(s)
- Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yu Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Yilong Huang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lanhong Guo
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chunyu Chang
- College of Chemistry and Molecular Sciences, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, and Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan, Hubei, 430072, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
11
|
Nie MD, Li N, Huang ZB, Cheng RS, Zhang Q, Fu LJ, Cheng CK. Innovative hydrogel-patch combination for large annulus fibrosus defects: a prospective approach to address herniation recurrence. Spine J 2024; 24:2002-2012. [PMID: 38914373 DOI: 10.1016/j.spinee.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND CONTEXT Large annulus fibrosus (AF) defects often lead to a high rate of reherniation, particularly in the medial AF region, which has limited self-healing capabilities. The increasing prevalence of herniated discs underscores the need for effective repair strategies. PURPOSE The objectives of this study were to design an AF repair technique to reduce solve the current problems of insufficient mechanical properties and poor sealing capacity. STUDY DESIGN In vitro biomechanical experiments and finite element analysis. METHODS The materials used in this study were patches and hydrogels with good biocompatibility and sufficient mechanical properties to withstand loading in the lumbar spine. Five repair techniques were assessed in this study: hydrogel filler (HF), AF patch medial barrier (MB), AF patch medial barrier and hydrogel filler (MB&HF), AF patch medial-lateral barrier (MLB), and AF patch medial-lateral barrier and hydrogel filler (MLB&HF). The repair techniques were subjected to in vitro testing (400 N axial compression and 0-500 N fatigue loading at 5Hz) and finite element analysis (400 N axial compression) to evaluate the effectiveness at repairing large AF defects. The evaluation included repair tightness, spinal stability, and fatigue resistance. RESULTS From the in vitro testing, the failure load of the repair techniques was in the following order HF MLB >MB&HF >MLB&HF. CONCLUSIONS The combined use of patches and hydrogels exhibited promising mechanical properties postdiscectomy, providing a promising solution for addressing large AF defects and improving disc stability. CLINICAL SIGNIFICANCE This study introduces a promising method for repairing large annular fissure (AF) defects after disc herniation, combining patch repair with a hydrogel filler. These techniques hold potential for developing clinical AF repair products to address this challenging issue.
Collapse
Affiliation(s)
- Mao-Dan Nie
- School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai, 200030, China
| | - Na Li
- Department of Absorbable Materials Research and Development, Beijing Naton Medical Institute, No.9 Chengwan Street, Haidian District, Beijing, 100143, China
| | - Ze-Bin Huang
- Department of Spine Surgery, First Affiliated Hospital of Second Military Medical University, No. 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Rong-Shan Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai, 200030, China
| | - Qiang Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai, 200030, China
| | - Ling-Jie Fu
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Manufacturing Bureau Road, Huangpu District, Shanghai, 200011, China
| | - Cheng-Kung Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai, 200030, China.
| |
Collapse
|
12
|
Kleiner S, Wulf V, Bisker G. Single-walled carbon nanotubes as near-infrared fluorescent probes for bio-inspired supramolecular self-assembled hydrogels. J Colloid Interface Sci 2024; 670:439-448. [PMID: 38772260 DOI: 10.1016/j.jcis.2024.05.098] [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: 02/04/2024] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
Hydrogels derived from fluorenylmethoxycarbonyl (Fmoc)-conjugated amino acids and peptides demonstrate remarkable potential in biomedical applications, including drug delivery, tissue regeneration, and tissue engineering. These hydrogels can be injectable, offering a minimally invasive approach to hydrogel implantation. Given their potential for prolonged application, there is a need for non-destructive evaluation of their properties over extended periods. Thus, we introduce a hydrogel characterization platform employing single-walled carbon nanotubes (SWCNTs) as near-infrared (NIR) fluorescent probes. Our approach involves generating supramolecular self-assembling hydrogels from aromatic Fmoc-amino acids. Integrating SWCNTs into the hydrogels maintains their structural and mechanical properties, establishing SWCNTs as optical probes for hydrogels. We demonstrate that the SWCNT NIR-fluorescence changes during the gelation process correlate to rheological changes within the hydrogels. Additionally, single particle tracking of SWCNTs incorporated in the hydrogels provides insights into differences in hydrogel morphologies. Furthermore, the disassembly process of the hydrogels can be monitored through the SWCNT fluorescence modulation. The unique attribute of SWCNTs as non-photobleaching fluorescent sensors, emitting at the biologically transparent window, offers a non-destructive method for studying hydrogel dynamics over extended periods. This platform could be applied to a wide range of self-assembling hydrogels to advance our understanding and applications of supramolecular assembly technologies.
Collapse
Affiliation(s)
- Shirel Kleiner
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Verena Wulf
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gili Bisker
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel; Center for Light-Matter Interaction, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
13
|
Rijns L, Rutten MGTA, Vrehen AF, Aldana AA, Baker MB, Dankers PYW. Mimicking the extracellular world: from natural to fully synthetic matrices utilizing supramolecular biomaterials. NANOSCALE 2024; 16:16290-16312. [PMID: 39161293 DOI: 10.1039/d4nr02088j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
The extracellular matrix (ECM) has evolved around complex covalent and non-covalent interactions to create impressive function-from cellular signaling to constant remodeling. A major challenge in the biomedical field is the de novo design and control of synthetic ECMs for applications ranging from tissue engineering to neuromodulation to bioelectronics. As we move towards recreating the ECM's complexity in hydrogels, the field has taken several approaches to recapitulate the main important features of the native ECM (i.e. mechanical, bioactive and dynamic properties). In this review, we first describe the wide variety of hydrogel systems that are currently used, ranging from fully natural to completely synthetic to hybrid versions, highlighting the advantages and limitations of each class. Then, we shift towards supramolecular hydrogels that show great potential for their use as ECM mimics due to their biomimetic hierarchical structure, inherent (controllable) dynamic properties and their modular design, allowing for precise control over their mechanical and biochemical properties. In order to make the next step in the complexity of synthetic ECM-mimetic hydrogels, we must leverage the supramolecular self-assembly seen in the native ECM; we therefore propose to use supramolecular monomers to create larger, hierarchical, co-assembled hydrogels with complex and synergistic mechanical, bioactive and dynamic features.
Collapse
Affiliation(s)
- Laura Rijns
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Martin G T A Rutten
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Annika F Vrehen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Ana A Aldana
- Department of Complex Tissue Regeneration, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Matthew B Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| |
Collapse
|
14
|
Yang S, Zhu Y, Shi Y, Su S, Liang H, Li S, Wu Z, Miao J, Chen Y, Zhang X, Wang X. Screening of NSAIDs library identifies Tinoridine as a novel ferroptosis inhibitor for potential intervertebral disc degeneration therapy. Free Radic Biol Med 2024; 221:245-256. [PMID: 38806104 DOI: 10.1016/j.freeradbiomed.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Low back pain (LBP) may profoundly impact the quality of life across the globe, and intervertebral disc degeneration (IVDD) is the major cause of LBP; however, targeted pharmaceutical interventions for IVDD are still lacking. Ferroptosis is a novel form of iron-dependent programmed cell death. Studies have showed that ferroptosis may closely associate with IVDD; thus, targeting ferroptosis may have great potential for IVDD therapy. Non-steroidal anti-inflammatory drugs (NSAIDs) are the first-line medications for LBP, while nuclear factor-erythroid 2-related factor-2 (Nrf2) is a key inhibitory protein for ferroptosis. In the current study, we conducted a molecular docking screening between NSAIDs library and Nrf2 protein. Tinoridine was shown to have a high binding affinity to Nrf2. The in vitro study in nucleus pulposus (NP) cells showed that Tinoridine may promote the expression and activity of Nrf2, it may also rescue RSL3-induced ferroptosis in NP cells. Knockdown of Nrf2 reverses the protective effect of Tinoridine on RSL3-induced ferroptosis in NP cells, suggesting that the inhibitory effect of Tinoridine on ferroptosis is through Nrf2. In vivo study demonstrated that Tinoridine may attenuate the progression of IVDD in rats. As NSAIDs are already clinically used for LBP therapy, the current study supports Tinoridine's application from the view of ferroptosis inhibition.
Collapse
Affiliation(s)
- Shu Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuxuan Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shenkai Su
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Haibo Liang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Sunlong Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhouwei Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiansen Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuli Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
15
|
Rijns L, Baker MB, Dankers PYW. Using Chemistry To Recreate the Complexity of the Extracellular Matrix: Guidelines for Supramolecular Hydrogel-Cell Interactions. J Am Chem Soc 2024; 146:17539-17558. [PMID: 38888174 PMCID: PMC11229007 DOI: 10.1021/jacs.4c02980] [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: 02/28/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Hydrogels have emerged as a promising class of extracellular matrix (ECM)-mimicking materials in regenerative medicine. Here, we briefly describe current state-of-the-art of ECM-mimicking hydrogels, ranging from natural to hybrid to completely synthetic versions, giving the prelude to the importance of supramolecular interactions to make true ECM mimics. The potential of supramolecular interactions to create ECM mimics for cell culture is illustrated through a focus on two different supramolecular hydrogel systems, both developed in our laboratories. We use some recent, significant findings to present important design principles underlying the cell-material interaction. To achieve cell spreading, we propose that slow molecular dynamics (monomer exchange within fibers) is crucial to ensure the robust incorporation of cell adhesion ligands within supramolecular fibers. Slow bulk dynamics (stress-relaxation─fiber rearrangements, τ1/2 ≈ 1000 s) is required to achieve cell spreading in soft gels (<1 kPa), while gel stiffness overrules dynamics in stiffer gels. Importantly, this resonates with the findings of others which specialize in different material types: cell spreading is impaired in case substrate relaxation occurs faster than clutch binding and focal adhesion lifetime. We conclude with discussing considerations and limitations of the supramolecular approach as well as provide a forward thinking perspective to further understand supramolecular hydrogel-cell interactions. Future work may utilize the presented guidelines underlying cell-material interactions to not only arrive at the next generation of ECM-mimicking hydrogels but also advance other fields, such as bioelectronics, opening up new opportunities for innovative applications.
Collapse
Affiliation(s)
- Laura Rijns
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Matthew B. Baker
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology
Inspired Regenerative Medicine, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - Patricia Y. W. Dankers
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Chemical Engineering and Chemistry, Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
16
|
Omidian H, Chowdhury SD, Wilson RL. Advancements and Challenges in Hydrogel Engineering for Regenerative Medicine. Gels 2024; 10:238. [PMID: 38667657 PMCID: PMC11049258 DOI: 10.3390/gels10040238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
This manuscript covers the latest advancements and persisting challenges in the domain of tissue engineering, with a focus on the development and engineering of hydrogel scaffolds. It highlights the critical role of these scaffolds in emulating the native tissue environment, thereby providing a supportive matrix for cell growth, tissue integration, and reducing adverse reactions. Despite significant progress, this manuscript emphasizes the ongoing struggle to achieve an optimal balance between biocompatibility, biodegradability, and mechanical stability, crucial for clinical success. It also explores the integration of cutting-edge technologies like 3D bioprinting and biofabrication in constructing complex tissue structures, alongside innovative materials and techniques aimed at enhancing tissue growth and functionality. Through a detailed examination of these efforts, the manuscript sheds light on the potential of hydrogels in advancing regenerative medicine and the necessity for multidisciplinary collaboration to navigate the challenges ahead.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.D.C.); (R.L.W.)
| | | | | |
Collapse
|
17
|
Li ZL, Lu Q, Honiball JR, Wan SHT, Yeung KWK, Cheung KMC. Mechanical characterization and design of biomaterials for nucleus pulposus replacement and regeneration. J Biomed Mater Res A 2023; 111:1888-1902. [PMID: 37555381 DOI: 10.1002/jbm.a.37593] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/10/2023]
Abstract
Biomaterials for nucleus pulposus (NP) replacement and regeneration have great potential to restore normal biomechanics in degenerated intervertebral discs following nucleotomy. Mechanical characterizations are essential for assessing the efficacy of biomaterial implants for clinical applications. While traditional compression tests are crucial to quantify various modulus values, relaxation behaviors and fatigue resistance, rheological measurements should also be conducted to investigate the viscoelastic properties, injectability, and overall stability upon deformation. To recapitulate the physiological in vivo environment, the use of spinal models is necessary to evaluate the risk of implant extrusion and the restoration of biomechanics under different loading conditions. When designing devices for NP replacement, injectable materials are ideal to fully fill the nucleus cavity and prevent implant migration. In addition to achieving biocompatibility and desirable mechanical characteristics, biomaterial implants should be optimized to avoid implant extrusion or re-herniation post-operatively. This review discusses the most commonly used testing protocols for assessing mechanical properties of biomaterial implants and serves as reference material for enabling researchers to characterize NP implants through a unified approach whereby newly developed biomaterials may be compared and contrasted to existing devices.
Collapse
Affiliation(s)
- Zhuoqi Lucas Li
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Qiuji Lu
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - John Robert Honiball
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Sandra Hiu-Tung Wan
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Kelvin Wai-Kwok Yeung
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Kenneth Man-Chee Cheung
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
18
|
Han H, Zhao X, Ma H, Zhang Y, Lei B. Multifunctional injectable hydrogels with controlled delivery of bioactive factors for efficient repair of intervertebral disc degeneration. Heliyon 2023; 9:e21867. [PMID: 38027562 PMCID: PMC10665751 DOI: 10.1016/j.heliyon.2023.e21867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/07/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Millions of people worldwide suffer from intervertebral disc degeneration (IVDD), which imposes a significant socioeconomic burden on society. There is an urgent clinical demand for more effective treatments for IVDD because conventional treatments can only alleviate the symptoms rather than preventing the progression of IVDD. Hydrogels, a class of elastic biomaterials with good biocompatibility, are promising candidates for intervertebral disc repair and regeneration. In recent years, various hydrogels have been investigated in vitro and in vivo for the repair of intervertebral discs, some of which are ready for clinical testing. This review summarizes the latest findings and developments in using bioactive factors-released bioactive injectable hydrogels for the repair and regeneration of intervertebral discs. It focuses on the analysis and summary of the use of multifunctional injectable hydrogels to delivery bioactive factors (cells, exosomes, growth factors, genes, drugs) for disc regeneration, providing guidance for future study. Finally, we discussed and analyzed the optimal timing for the application of controlled-release hydrogels in the treatment of IVDD to meet the high standards required for intervertebral disc regeneration and precision medicine.
Collapse
Affiliation(s)
- Hao Han
- Department of Orthopaedics of the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaoming Zhao
- Department of Orthopaedics of the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongyun Ma
- Department of Orthopaedics of the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yingang Zhang
- Department of Orthopaedics of the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Bo Lei
- Department of Orthopaedics of the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710000, China
- Fronter Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
| |
Collapse
|
19
|
Zhang J, Zhao D, Lu K. Mechanisms and influencing factors of peptide hydrogel formation and biomedicine applications of hydrogels. SOFT MATTER 2023; 19:7479-7493. [PMID: 37756117 DOI: 10.1039/d3sm01057k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Self-assembled peptide-based hydrogels have shown great potential in bio-related applications due to their porous structure, strong mechanical stability, high biocompatibility, and easy functionalization. Herein, the structure and characteristics of hydrogels and the mechanism of action of several regular secondary structures during gelation are investigated. The factors influencing the formation of peptide hydrogels, especially the pH responsiveness and salt ion induction are analyzed and summarized. Finally, the biomedical applications of peptide hydrogels, such as bone tissue engineering, cell culture, antigen presentation, antibacterial materials, and drug delivery are reviewed.
Collapse
Affiliation(s)
- Jiahui Zhang
- School of Chemistry and Chemical Engineering, Henan University of Technology, Locus Street, High-Tech Industry Development Zone, Zhengzhou 450001, China.
| | - Dongxin Zhao
- School of Chemistry and Chemical Engineering, Henan University of Technology, Locus Street, High-Tech Industry Development Zone, Zhengzhou 450001, China.
| | - Kui Lu
- School of Chemistry and Chemical Engineering, Henan University of Technology, Locus Street, High-Tech Industry Development Zone, Zhengzhou 450001, China.
- School of Chemical Engineering and Food Science, Zhengzhou University of Technology, Yingcai Road 18, Zhengzhou, 450044, Henan Province, China.
| |
Collapse
|
20
|
McFetridge ML, Kulkarni K, Lee TH, Del Borgo MP, Aguilar MI, Ricardo SD. Elucidating the cell penetrating properties of self-assembling β-peptides. NANOSCALE 2023; 15:14971-14980. [PMID: 37661822 DOI: 10.1039/d3nr03673a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Self-assembling lipopeptide hydrogels have been widely developed for the delivery of therapeutics due to their rapid gelation, injectability, and highly controlled physicochemical properties. Lipopeptides are also known for their membrane-associating and cell penetrating properties, which may impact on their application in cell-encapsulation. Self-assembling lipidated-β3-peptide materials developed in our laboratory have previously been used in cell culture as 2D substrates, thus as a continuation of this work we aimed to encapsulate cells in 3D by forming a hydrogel. We therefore assessed the self-assembling lipidated-β3-peptides for cell-penetrating properties in mesenchymal stems cells (MSC) using fluorescence microscopy and membrane association with surface plasmon resonance spectroscopy (SPR). The results demonstrated that lipidated β3-peptides penetrate the MSC plasma membrane and localise to the mitochondrial network. While self-assembling lipopeptide hydrogels have shown tremendous potential for delivery of therapeutics, further optimisation may be required to minimise the membrane uptake of the lipidated-β3-peptides for cell encapsulation applications.
Collapse
Affiliation(s)
- Meg L McFetridge
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Tzong-Hsien Lee
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Mark P Del Borgo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
21
|
Lv J, Wu Y, Cao Z, Liu X, Sun Y, Zhang P, Zhang X, Tang K, Cheng M, Yao Q, Zhu Y. Enhanced Cartilage and Subchondral Bone Repair Using Carbon Nanotube-Doped Peptide Hydrogel-Polycaprolactone Composite Scaffolds. Pharmaceutics 2023; 15:2145. [PMID: 37631359 PMCID: PMC10458387 DOI: 10.3390/pharmaceutics15082145] [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: 07/14/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
A carbon nanotube-doped octapeptide self-assembled hydrogel (FEK/C) and a hydrogel-based polycaprolactone PCL composite scaffold (FEK/C3-S) were developed for cartilage and subchondral bone repair. The composite scaffold demonstrated modulated microstructure, mechanical properties, and conductivity by adjusting CNT concentration. In vitro evaluations showed enhanced cell proliferation, adhesion, and migration of articular cartilage cells, osteoblasts, and bone marrow mesenchymal stem cells. The composite scaffold exhibited good biocompatibility, low haemolysis rate, and high protein absorption capacity. It also promoted osteogenesis and chondrogenesis, with increased mineralization, alkaline phosphatase (ALP) activity, and glycosaminoglycan (GAG) secretion. The composite scaffold facilitated accelerated cartilage and subchondral bone regeneration in a rabbit knee joint defect model. Histological analysis revealed improved cartilage tissue formation and increased subchondral bone density. Notably, the FEK/C3-S composite scaffold exhibited the most significant cartilage and subchondral bone formation. The FEK/C3-S composite scaffold holds great promise for cartilage and subchondral bone repair. It offers enhanced mechanical support, conductivity, and bioactivity, leading to improved tissue regeneration. These findings contribute to the advancement of regenerative strategies for challenging musculoskeletal tissue defects.
Collapse
Affiliation(s)
- Jiayi Lv
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xu Liu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xin Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Kexin Tang
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Min Cheng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
22
|
Li S, Yu Q, Li H, Chen M, Jin Y, Liu D. Self-Assembled Peptide Hydrogels in Regenerative Medicine. Gels 2023; 9:653. [PMID: 37623108 PMCID: PMC10453854 DOI: 10.3390/gels9080653] [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: 07/18/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Regenerative medicine is a complex discipline that is becoming a hot research topic. Skin, bone, and nerve regeneration dominate current treatments in regenerative medicine. A new type of drug is urgently needed for their treatment due to their high vulnerability to damage and weak self-repairing ability. A self-assembled peptide hydrogel is a good scaffolding material in regenerative medicine because it is similar to the cytoplasmic matrix environment; it promotes cell adhesion, migration, proliferation, and division; and its degradation products are natural and harmless proteins. However, fewer studies have examined the specific mechanisms of self-assembled peptide hydrogels in promoting tissue regeneration. This review summarizes the applications and mechanisms of self-assembled short peptide and peptide hydrogels in skin, bone, and neural healing to improve their applications in tissue healing and regeneration.
Collapse
Affiliation(s)
- Shuangyang Li
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.L.); (Q.Y.); (H.L.); (M.C.)
| | - Qixuan Yu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.L.); (Q.Y.); (H.L.); (M.C.)
| | - Hongpeng Li
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.L.); (Q.Y.); (H.L.); (M.C.)
| | - Meiqi Chen
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.L.); (Q.Y.); (H.L.); (M.C.)
| | - Ye Jin
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.L.); (Q.Y.); (H.L.); (M.C.)
| |
Collapse
|
23
|
Chen X, Kohan S, Bhargav D, Choi J, Perera S, Dean C, Chopra N, Sial A, Sandhu HS, Apos E, Appleyard R, Diwan AD. Phase 1 evaluation of an elastomeric nucleus pulposus device as an option to augment disc at microdiscectomy: Experimental results from biomechanical and biocompatibility testing and first in human. JOR Spine 2023; 6:e1250. [PMID: 37361335 PMCID: PMC10285756 DOI: 10.1002/jsp2.1250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 06/28/2023] Open
Abstract
Objective Whilst microdiscectomy is an excellent reliever of pain for recalcitrant lumbar disc herniation (LDH), it has a high failure rate over time due to the ensuing reduction in mechanical stabilization and support of the spine. One option is to clear the disc and replace it with a nonhygroscopic elastomer. Here, we present the evaluation of biomechanical and biological behavior of a novel elastomeric nucleus device (Kunovus disc device [KDD]), consisting of a silicone jacket and a two-part in situ curing silicone polymer filler. Materials and Methods ISO 10993 and American Society for Testing and Materials (ASTM) standards were used to evaluate the biocompatibility and mechanics of KDD. Sensitization, intracutaneous reactivity, acute systemic toxicity, genotoxicity, muscle implantation study, direct contact matrix toxicity assay, and cell growth inhibition assay were performed. Fatigue test, static compression creep testing, expulsion testing, swell testing, shock testing, and aged fatigue testing were conducted to characterize the mechanical and wear behavior of the device. Cadaveric studies to develop a surgical manual and evaluate feasibility were conducted. Finally, a first-in-human implantation was conducted to complete the proof of principle. Results The KDD demonstrated exceptional biocompatibility and biodurability. Mechanical tests showed no Barium-containing particles in fatigue test, no fracture of nucleus in static compression creep testing, no extrusion and swelling, and no material failure in shock and aged fatigue testing. Cadaver training sessions showed that KDD was deemed implantable during microdiscectomy procedures in a minimally invasive manner. Following IRB approval, the first implantation in a human showed no intraoperative vascular and neurological complications and demonstrated feasibility. This successfully completed Phase 1 development of the device. Conclusion The elastomeric nucleus device may mimic native disc behavior in mechanical tests, offering an effective way for treating LDH by way of Phase 2 and subsequent clinical trials or postmarket surveillance in the future.
Collapse
Affiliation(s)
- Xiaolong Chen
- Spine Labs, St. George & Sutherland Clinical School, University of New South WalesSydneyNew South WalesAustralia
- Kunovus TechnologiesSydneyNew South WalesAustralia
| | - Saeed Kohan
- St. George Hospital, University of New South WalesSydneyNew South WalesAustralia
| | | | | | | | - Cameron Dean
- Kunovus TechnologiesSydneyNew South WalesAustralia
| | - Neha Chopra
- Spine Labs, St. George & Sutherland Clinical School, University of New South WalesSydneyNew South WalesAustralia
- Spine Service, Department of Orthopaedic SurgerySt. George Hospital CampusSydneyNew South WalesAustralia
| | - Alisha Sial
- Spine Labs, St. George & Sutherland Clinical School, University of New South WalesSydneyNew South WalesAustralia
- Spine Service, Department of Orthopaedic SurgerySt. George Hospital CampusSydneyNew South WalesAustralia
| | - Harvinder S. Sandhu
- Spinal Surgical Service, Hospital for Special Surgery, Weill Medical College of Cornell UniversityNew YorkNew YorkUSA
| | - Esther Apos
- Kunovus TechnologiesSydneyNew South WalesAustralia
- Cmsscidoc Pty LtdMelbourneVictoriaAustralia
| | - Richard Appleyard
- Orthopaedic Biomechanics Research Group, Department of Biomedical Sciences, Faculty of Medicine and Health SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Ashish D. Diwan
- Spine Labs, St. George & Sutherland Clinical School, University of New South WalesSydneyNew South WalesAustralia
- Spine Service, Department of Orthopaedic SurgerySt. George Hospital CampusSydneyNew South WalesAustralia
| |
Collapse
|
24
|
Treherne JM, Miller AF. Novel hydrogels: are they poised to transform 3D cell-based assay systems in early drug discovery? Expert Opin Drug Discov 2023; 18:335-346. [PMID: 36722285 DOI: 10.1080/17460441.2023.2175813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Success in drug discovery remains unpredictable. However, more predictive and relevant disease models are becoming pivotal to demonstrating the clinical benefits of new drugs earlier in the lengthy drug discovery process. Novel hydrogel scaffolds are being developed to transform the relevance of such 3D cell-based in vitro assay systems. AREAS COVERED Most traditional hydrogels are still of unknown composition and suffer significant batch-to-batch variations, which lead to technical constraints. This article looks at how a new generation of novel synthetic hydrogels that are based on self-assembling peptides are poised to transform 3D cell-based assay systems by improving their relevance, reproducibility and scalability. EXPERT OPINION The emerging advantages of using these novel hydrogels for human 3D screening assays should enable the discovery of more cost-effective drugs, leading to improved patient benefits. Such a disruptive change could also reduce the considerable time lag from obtaining in vitro assay data to initiating clinical trials. There is now a sufficient body of data available in the literature to enable this ambition to become a reality by significantly improving the predictive validity of 3D cell-based assays in early drug discovery. Novel hydrogels are key to unlocking the full potential of these assay systems.
Collapse
Affiliation(s)
- J Mark Treherne
- Talisman Therapeutics Ltd, Jonas Webb Building and Cell Guidance Sysyems Ltd, Babraham Research Campus, Cambridge, UK
| | - Aline F Miller
- Manchester Institute of Biotechnology, School of Engineering, The University of Manchester, Oxford Road, Manchester, UK
| |
Collapse
|
25
|
Treacy NJ, Clerkin S, Davis JL, Kennedy C, Miller AF, Saiani A, Wychowaniec JK, Brougham DF, Crean J. Growth and differentiation of human induced pluripotent stem cell (hiPSC)-derived kidney organoids using fully synthetic peptide hydrogels. Bioact Mater 2023; 21:142-156. [PMID: 36093324 PMCID: PMC9420433 DOI: 10.1016/j.bioactmat.2022.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/27/2022] [Accepted: 08/01/2022] [Indexed: 11/15/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived kidney organoids have prospective applications ranging from basic disease modelling to personalised medicine. However, there remains a necessity to refine the biophysical and biochemical parameters that govern kidney organoid formation. Differentiation within fully-controllable and physiologically relevant 3D growth environments will be critical to improving organoid reproducibility and maturation. Here, we matured hiPSC-derived kidney organoids within fully synthetic self-assembling peptide hydrogels (SAPHs) of variable stiffness (storage modulus, G'). The resulting organoids contained complex structures comparable to those differentiated within the animal-derived matrix, Matrigel. Single-cell RNA sequencing (scRNA-seq) was then used to compare organoids matured within SAPHs to those grown within Matrigel or at the air-liquid interface. A total of 13,179 cells were analysed, revealing 14 distinct clusters. Organoid compositional analysis revealed a larger proportion of nephron cell types within Transwell-derived organoids, while SAPH-derived organoids were enriched for stromal-associated cell populations. Notably, differentiation within a higher G' SAPH generated podocytes with more mature gene expression profiles. Additionally, maturation within a 3D microenvironment significantly reduced the derivation of off-target cell types, which are a known limitation of current kidney organoid protocols. This work demonstrates the utility of synthetic peptide-based hydrogels with a defined stiffness, as a minimally complex microenvironment for the selected differentiation of kidney organoids.
Collapse
Affiliation(s)
- Niall J Treacy
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Shane Clerkin
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Jessica L Davis
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Ciarán Kennedy
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| | - Aline F Miller
- Department of Materials & Manchester Institute of Biotechnology (MIB), School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Alberto Saiani
- Department of Materials & Manchester Institute of Biotechnology (MIB), School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Jacek K Wychowaniec
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dermot F Brougham
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - John Crean
- Diabetes Complications Research Centre, University College Dublin (UCD) Conway Institute of Biomolecular and Biomedical Research and Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, Belfield, Dublin 4, Ireland
| |
Collapse
|
26
|
Poirier A, Le Griel P, Bizien T, Zinn T, Pernot P, Baccile N. Shear recovery and temperature stability of Ca 2+ and Ag + glycolipid fibrillar metallogels with unusual β-sheet-like domains. SOFT MATTER 2023; 19:366-377. [PMID: 36508178 DOI: 10.1039/d2sm00374k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Low-molecular weight gelators (LMWGs) are small molecules (Mw < ∼1 kDa), which form self-assembled fibrillar network (SAFiN) hydrogels in water. A great majority of SAFiN gels are described by an entangled network of self-assembled fibers, in analogy to a polymer in a good solvent. Here, fibrillation of a biobased glycolipid bolaamphiphile is triggered by Ca2+ or Ag+ ions which are added to its diluted micellar phase. The resulting SAFiN, which forms a hydrogel above 0.5 wt%, has a "nano-fishnet" structure, characterized by a fibrous network of both entangled fibers and β-sheet-like rafts, generally observed for silk fibroin, actin hydrogels or mineral imogolite nanotubes, but generally not known for SAFiN. This work focuses on the strength of the SAFIN gels, their fast recovery after applying a mechanical stimulus (strain) and their unusual resistance to temperature, studied by coupling rheology to small angle X-ray scattering (rheo-SAXS) using synchrotron radiation. The Ca2+-based hydrogel maintains its properties up to 55 °C, while the Ag+-based gel shows a constant elastic modulus up to 70 °C, without the appearance of any gel-to-sol transition temperature. Furthermore, the glycolipid is obtained by fermentation from natural resources (glucose and rapeseed oil), thus showing that naturally engineered compounds can have unprecedented properties, when compared to the wide range of chemically derived amphiphiles.
Collapse
Affiliation(s)
- Alexandre Poirier
- Sorbonne Université, Centre National de la Recherche Scientifique, Laboratoire de Chimie de la Matière Condensée de Paris, LCMCP, F-75005 Paris, France.
| | - Patrick Le Griel
- Sorbonne Université, Centre National de la Recherche Scientifique, Laboratoire de Chimie de la Matière Condensée de Paris, LCMCP, F-75005 Paris, France.
| | - Thomas Bizien
- Synchrotron Soleil, L'Orme des Merisiers, Saint-Aubin, BP48, 91192 Gif-sur-Yvette Cedex, France
| | - Thomas Zinn
- ESRF - The European Synchrotron, CS40220, 38043 Grenoble, France
| | - Petra Pernot
- ESRF - The European Synchrotron, CS40220, 38043 Grenoble, France
| | - Niki Baccile
- Sorbonne Université, Centre National de la Recherche Scientifique, Laboratoire de Chimie de la Matière Condensée de Paris, LCMCP, F-75005 Paris, France.
| |
Collapse
|
27
|
Yadav N, Kumar U, Roopmani P, Krishnan UM, Sethuraman S, Chauhan MK, Chauhan VS. Ultrashort Peptide-Based Hydrogel for the Healing of Critical Bone Defects in Rabbits. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54111-54126. [PMID: 36401830 DOI: 10.1021/acsami.2c18733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The use of hydrogels as scaffolds for three-dimensional (3D) cell growth is an active area of research in tissue engineering. Herein, we report the self-assembly of an ultrashort peptide, a tetrapeptide, Asp-Leu-IIe-IIe, the shortest peptide sequence from a highly fibrillogenic protein TDP-43, into the hydrogel. The hydrogel was mechanically strong and highly stable, with storage modulus values in MPa ranges. The hydrogel supported the proliferation and successful differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) in its matrix as assessed by cell viability, calcium deposition, alkaline phosphatase (ALP) activity, and the expression of osteogenic marker gene studies. To check whether the hydrogel supports 3D growth and regeneration in in vivo conditions, a rabbit critical bone defect model was used. Micro-computed tomography (CT) and X-ray analysis demonstrated the formation of mineralized neobone in the defect areas, with significantly higher bone mineralization and relative bone densities in animals treated with the peptide hydrogel compared to nontreated and matrigel treatment groups. The ultrashort peptide-based hydrogel developed in this work holds great potential for its further development as tissue regeneration and/or engineering scaffolds.
Collapse
Affiliation(s)
- Nitin Yadav
- Molecular Medicine Group, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi110067, India
- Delhi Institute of Pharmaceutical Sciences and Research, Mehrauli-Badarpur Road, Sector-3, Pushpvihar, New Delhi110017, India
| | - Utkarsh Kumar
- Molecular Medicine Group, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi110067, India
| | - Purandhi Roopmani
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA's Hub for Research & Innovation (SHRI), School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur613401, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA's Hub for Research & Innovation (SHRI), School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur613401, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA's Hub for Research & Innovation (SHRI), School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur613401, India
| | - Meenakshi K Chauhan
- Delhi Institute of Pharmaceutical Sciences and Research, Mehrauli-Badarpur Road, Sector-3, Pushpvihar, New Delhi110017, India
| | - Virander S Chauhan
- Molecular Medicine Group, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi110067, India
| |
Collapse
|
28
|
Dong Z, Chen S, Wang L, Qi P, Wei L. Fabrication of Flower-stacked structured microparticles encapsulated with Stem cells and Growth Factor to the potential treatment of Intervertebral Disc Degeneration. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.04.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
29
|
Acidic and basic self-assembling peptide and peptide-graphene oxide hydrogels: characterisation and effect on encapsulated nucleus pulposus cells. Acta Biomater 2022; 143:145-158. [PMID: 35196554 DOI: 10.1016/j.actbio.2022.02.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 12/28/2022]
Abstract
Extracellular pH can have a profound effect on cell metabolism, gene and protein expression. Nucleus pulposus (NP) cells, for example, under acidic conditions accelerate the production of degradative enzymes and pro-inflammatory cytokines, leading ultimately to intervertebral disc degeneration, a major cause of back pain. Self-assembling peptide hydrogels constitute a well-established class of biomaterials that could be exploited as pH-tunable platform to investigate cell behaviour under normal and non-physiological pH. In this paper we formulated acidic (pH = 4) and basic (pH = 9) hydrogels, from the same octapeptide FEFKFEFK (F8) (F = phenyalanine, E = glutamic acid, K = lysine), to test the effect of non-physiological pH on encapsulated NP cells. Similarly, graphene oxide-containing F8 hydrogels (GO-F8) were formulated as stiffer analogues. Acidic and basic hydrogels showed peculiar morphologies and rheological properties, with all systems able to buffer within 30 minutes of exposure to cell culture media. NP cells seeded in acidic F8 hydrogels showed a more catabolic phenotype compared to basic hydrogels, with increased gene expression of degradative enzymes (MMP-3, ADAMTS-4), neurotrophic factors (NGF and BDNF) and NF-κB p65 phosphorylation. Acidic GO-F8 hydrogels also induced a catabolic response, although milder than basic counterparts and with the highest gene expression of characteristic NP-matrix components, aggrecan and collagen II. In all systems, the cellular response had a peak within 3 days of encapsulation, thereafter decreasing over 7 days, suggesting a 'transitory' effect of hydrogel pH on encapsulated cells. This work gives an insight on the effect of pH (and pH buffering) on encapsulated NP cells and offers new designs of low and high pH peptide hydrogels for 3D cell culture studies. STATEMENT OF SIGNIFICANCE: We have recently shown the potential of graphene oxide - self-assembling peptide hybrid hydrogels for NP cell culture and regeneration. Alongside cell carrier, self-assembling peptide hydrogels actually provide a versatile pH-tunable platform for biological studies. In this work we decided to explore the effect of non-physiological pH (and pH buffering) on encapsulated NP cells. Our approach allows the formulation of both acidic and basic hydrogels, starting from the same peptide sequence. We showed that the initial pH of the scaffold does not affect significantly cell response to encapsulation, but the presence of GO results in lower inflammatory levels and higher NP matrix protein production. This platform could be exploited to study the effect of pH on different cell types whose behaviour can be pH-dependent.
Collapse
|
30
|
Ligorio C, Hoyland JA, Saiani A. Self-Assembling Peptide Hydrogels as Functional Tools to Tackle Intervertebral Disc Degeneration. Gels 2022; 8:gels8040211. [PMID: 35448112 PMCID: PMC9028266 DOI: 10.3390/gels8040211] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022] Open
Abstract
Low back pain (LBP), caused by intervertebral disc (IVD) degeneration, is a major contributor to global disability. In its healthy state, the IVD is a tough and well-hydrated tissue, able to act as a shock absorber along the spine. During degeneration, the IVD is hit by a cell-driven cascade of events, which progressively lead to extracellular matrix (ECM) degradation, chronic inflammation, and pain. Current treatments are divided into palliative care (early stage degeneration) and surgical interventions (late-stage degeneration), which are invasive and poorly efficient in the long term. To overcome these limitations, alternative tissue engineering and regenerative medicine strategies, in which soft biomaterials are used as injectable carriers of cells and/or biomolecules to be delivered to the injury site and restore tissue function, are currently being explored. Self-assembling peptide hydrogels (SAPHs) represent a promising class of de novo synthetic biomaterials able to merge the strengths of both natural and synthetic hydrogels for biomedical applications. Inherent features, such as shear-thinning behaviour, high biocompatibility, ECM biomimicry, and tuneable physiochemical properties make these hydrogels appropriate and functional tools to tackle IVD degeneration. This review will describe the pathogenesis of IVD degeneration, list biomaterials requirements to attempt IVD repair, and focus on current peptide hydrogel materials exploited for this purpose.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M1 3BB, UK;
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PG, UK;
- Correspondence:
| | - Judith A. Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PG, UK;
| | - Alberto Saiani
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M1 3BB, UK;
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
| |
Collapse
|
31
|
Yi J, Liu Q, Zhang Q, Chew TG, Ouyang H. Modular protein engineering-based biomaterials for skeletal tissue engineering. Biomaterials 2022; 282:121414. [DOI: 10.1016/j.biomaterials.2022.121414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/27/2021] [Accepted: 05/19/2021] [Indexed: 12/24/2022]
|
32
|
Gray VP, Amelung CD, Duti IJ, Laudermilch EG, Letteri RA, Lampe KJ. Biomaterials via peptide assembly: Design, characterization, and application in tissue engineering. Acta Biomater 2022; 140:43-75. [PMID: 34710626 PMCID: PMC8829437 DOI: 10.1016/j.actbio.2021.10.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A core challenge in biomaterials, with both fundamental significance and technological relevance, concerns the rational design of bioactive microenvironments. Designed properly, peptides can undergo supramolecular assembly into dynamic, physical hydrogels that mimic the mechanical, topological, and biochemical features of native tissue microenvironments. The relatively facile, inexpensive, and automatable preparation of peptides, coupled with low batch-to-batch variability, motivates the expanded use of assembling peptide hydrogels for biomedical applications. Integral to realizing dynamic peptide assemblies as functional biomaterials for tissue engineering is an understanding of the molecular and macroscopic features that govern assembly, morphology, and biological interactions. In this review, we first discuss the design of assembling peptides, including primary structure (sequence), secondary structure (e.g., α-helix and β-sheets), and molecular interactions that facilitate assembly into multiscale materials with desired properties. Next, we describe characterization tools for elucidating molecular structure and interactions, morphology, bulk properties, and biological functionality. Understanding of these characterization methods enables researchers to access a variety of approaches in this ever-expanding field. Finally, we discuss the biological properties and applications of peptide-based biomaterials for engineering several important tissues. By connecting molecular features and mechanisms of assembling peptides to the material and biological properties, we aim to guide the design and characterization of peptide-based biomaterials for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Engineering peptide-based biomaterials that mimic the topological and mechanical properties of natural extracellular matrices provide excellent opportunities to direct cell behavior for regenerative medicine and tissue engineering. Here we review the molecular-scale features of assembling peptides that result in biomaterials that exhibit a variety of relevant extracellular matrix-mimetic properties and promote beneficial cell-biomaterial interactions. Aiming to inspire and guide researchers approaching this challenge from both the peptide biomaterial design and tissue engineering perspectives, we also present characterization tools for understanding the connection between peptide structure and properties and highlight the use of peptide-based biomaterials in neural, orthopedic, cardiac, muscular, and immune engineering applications.
Collapse
Affiliation(s)
- Vincent P Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Connor D Amelung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Israt Jahan Duti
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Emma G Laudermilch
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| |
Collapse
|
33
|
Culbert MP, Warren JP, Dixon AR, Fermor HL, Beales PA, Wilcox RK. Evaluation of injectable nucleus augmentation materials for the treatment of intervertebral disc degeneration. Biomater Sci 2021; 10:874-891. [PMID: 34951410 DOI: 10.1039/d1bm01589c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Back pain affects a person's health and mobility as well as being associated with large health and social costs. Lower back pain is frequently caused by degeneration of the intervertebral disc. Current operative and non-operative treatments are often ineffective and expensive. Nucleus augmentation is designed to be a minimally invasive method of restoring the disc to its native healthy state by restoring the disc height, and mechanical and/or biological properties. The majority of the candidate materials for nucleus augmentation are injectable hydrogels. In this review, we examine the materials that are currently under investigation for nucleus augmentation, and compare their ability to meet the design requirements for this application. Specifically, the delivery of the material into the disc, the mechanical properties of the material and the biological compatibility are examined. Recommendations for future testing are also made.
Collapse
Affiliation(s)
- Matthew P Culbert
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - James P Warren
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - Andrew R Dixon
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - Hazel L Fermor
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| | - Paul A Beales
- School of Chemistry, Astbury Centre for Structural Molecular Biology and Bragg Centre for Materials Research, University of Leeds, UK, LS2 9JT
| | - Ruth K Wilcox
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, UK, LS2 9JT.
| |
Collapse
|
34
|
Bao Y, Li Z, Li Y, Chen T, Cheng Y, Xu M. Recent Advances of Biomedical Materials for Prevention of Post-ESD Esophageal Stricture. Front Bioeng Biotechnol 2021; 9:792929. [PMID: 35004652 PMCID: PMC8727907 DOI: 10.3389/fbioe.2021.792929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Esophageal stricture commonly occurs in patients that have suffered from endoscopic submucosal dissection (ESD), and it makes swallowing difficult for patients, significantly reducing their life qualities. So far, the prevention strategies applied in clinical practice for post-ESD esophageal stricture usually bring various inevitable complications, which drastically counteract their effectiveness. Nowadays, with the widespread investigation and application of biomedical materials, lots of novel approaches have been devised in terms of the prevention of esophageal stricture. Biomedical polymers and biomedical-derived materials are the most used biomedical materials to prevent esophageal stricture after ESD. Both of biomedical polymers and biomedical-derived materials possess great physicochemical properties such as biocompatibility and biodegradability. Moreover, some biomedical polymers can be used as scaffolds to promote cell growth, and biomedical-derived materials have biological functions similar to natural organisms, so they are important in tissue engineering. In this review, we have summarized the current approaches for preventing esophageal stricture and put emphasis on the discussion of the roles biomedical polymers and biomedical-derived materials acted in esophageal stricture prevention. Meanwhile, we proposed several potential methods that may be highly rational and feasible in esophageal stricture prevention based on other researches associated with biomedical materials. This review is expected to offer a significant inspiration from biomedical materials to explore more effective, safer, and more economical strategies to manage post-ESD esophageal stricture.
Collapse
Affiliation(s)
- Yuchen Bao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhenguang Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingze Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Chen
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Cheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Meidong Xu
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Modeling multiaxial damage regional variation in human annulus fibrosus. Acta Biomater 2021; 136:375-388. [PMID: 34547514 DOI: 10.1016/j.actbio.2021.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023]
Abstract
In the present article, a fully three-dimensional human annulus fibrosus model is developed by considering the regional variation of the complex structural organization of collagen network at different scales to predict the regional anisotropic multiaxial damage of the intervertebral disc. The model parameters are identified using experimental data considering as elementary structural unit, the single annulus lamellae stretched till failure along the micro-sized collagen fibers. The multi-layered lamellar/inter-lamellar annulus model is constructed by considering the effective interactions between adjacent layers and the chemical-induced volumetric strain. The regional dependent model predictions are analyzed under various loading modes and compared to experimental data when available. The stretching along the circumferential and radial directions till failure serves to check the predictive capacities of the annulus model. Model results under simple shear, biaxial stretching and plane-strain compression are further presented and discussed. Finally, a full disc model is constructed using the regional annulus model and simulations are presented to assess the most likely failed areas under disc axial compression. STATEMENT OF SIGNIFICANCE: The damage in annulus soft tissues is a complex multiscale phenomenon due to a complex structural arrangement of collagen network at different scales of hierarchical organization. A fully three-dimensional constitutive representation that considers the regional variation of the structural complexity to estimate annulus multiaxial mechanics till failure has not yet been developed. Here, a model is developed to predict deformation-induced damage and failure of annulus under multiaxial loading histories considering as time-dependent physical process both chemical-induced volumetric effects and damage accumulation. After model identification using single lamellae extracted from different disc regions, the model predictability is verified for various multiaxial elementary loading modes representative of the spine movement. The heterogeneous mechanics of a full human disc model is finally presented.
Collapse
|
36
|
La Manna S, Di Natale C, Onesto V, Marasco D. Self-Assembling Peptides: From Design to Biomedical Applications. Int J Mol Sci 2021; 22:12662. [PMID: 34884467 PMCID: PMC8657556 DOI: 10.3390/ijms222312662] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022] Open
Abstract
Self-assembling peptides could be considered a novel class of agents able to harvest an array of micro/nanostructures that are highly attractive in the biomedical field. By modifying their amino acid composition, it is possible to mime several biological functions; when assembled in micro/nanostructures, they can be used for a variety of purposes such as tissue regeneration and engineering or drug delivery to improve drug release and/or stability and to reduce side effects. Other significant advantages of self-assembled peptides involve their biocompatibility and their ability to efficiently target molecular recognition sites. Due to their intrinsic characteristics, self-assembled peptide micro/nanostructures are capable to load both hydrophobic and hydrophilic drugs, and they are suitable to achieve a triggered drug delivery at disease sites by inserting in their structure's stimuli-responsive moieties. The focus of this review was to summarize the most recent and significant studies on self-assembled peptides with an emphasis on their application in the biomedical field.
Collapse
Affiliation(s)
- Sara La Manna
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Concetta Di Natale
- Istituto Italiano di Tecnologia, IIT@CRIB, Largo Barsanti e Matteucci, 53, 80125 Napoli, Italy
- Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, Università di Napoli Federico II, Piazzale Tecchio, 80, 80125 Napoli, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, Consiglio Nazionale delle Ricerche, CNR NANOTEC, via Monteroni, c/o Campus Ecotekne, 73100 Lecce, Italy;
| | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy;
| |
Collapse
|
37
|
Wang F, Gnewou O, Wang S, Osinski T, Zuo X, Egelman EH, Conticello VP. Deterministic chaos in the self-assembly of β sheet nanotubes from an amphipathic oligopeptide. MATTER 2021; 4:3217-3231. [PMID: 34632372 PMCID: PMC8494133 DOI: 10.1016/j.matt.2021.06.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The self-assembly of designed peptides into filaments and other higher-order structures has been the focus of intense interest because of the potential for creating new biomaterials and biomedical devices. These peptide assemblies have also been used as models for understanding biological processes, such as the pathological formation of amyloid. We investigate the assembly of an octapeptide sequence, Ac-FKFEFKFE-NH2, motivated by prior studies that demonstrated that this amphipathic β strand peptide self-assembled into fibrils and biocompatible hydrogels. Using high-resolution cryoelectron microscopy (cryo-EM), we are able to determine the atomic structure for two different coexisting forms of the fibrils, containing four and five β sandwich protofilaments, respectively. Surprisingly, the inner walls in both forms are parallel β sheets, while the outer walls are antiparallel β sheets. Our results demonstrate the chaotic nature of peptide self-assembly and illustrate the importance of cryo-EM structural analysis to understand the complex phase behavior of these materials at near-atomic resolution.
Collapse
Affiliation(s)
- Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Ordy Gnewou
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Shengyuan Wang
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Tomasz Osinski
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Xiaobing Zuo
- X-ray Science Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
- Correspondence: (E.H.E.), (V.P.C.)
| | - Vincent P. Conticello
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
- The Robert P. Apkarian Integrated Electron Microscopy Core (IEMC), Emory University, Atlanta, GA 30322, USA
- Lead contact
- Correspondence: (E.H.E.), (V.P.C.)
| |
Collapse
|
38
|
Qian Y, Di S, Wang L, Li Z. Recent advances in the synthesis and applications of graphene-polypeptide nanocomposites. J Mater Chem B 2021; 9:6521-6535. [PMID: 34318859 DOI: 10.1039/d1tb00779c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The combination of peptides and graphene-derived materials provides a new way to prepare graphene-based nanocomposites with unique structures, properties, and functions. The modification of graphene with different polypeptides not only improves the biocompatibility and biological recognition ability of graphene-based materials, but also greatly expands their application fields. In this work, we summarize different interactions between graphene and polypeptides, and the synthesis methods of novel functional graphene-polypeptide nanocomposites based on the interactions in recent years (from 2016 to present). In addition, the potential applications of graphene-peptide hybrid nanocomposites in biomedicine, tissue engineering, biosensors, environmental science engineering, optoelectronic materials, and energy storage are introduced. We hope that this review will help readers to understand the methods and mechanisms of the modification of graphene surfaces with biomolecules, and promote readers to understand the synthesis and applications of graphene-based nanocomposites. This work may provide hints and references for the development of peptide sequence design, and biomedical and functional materials, and will help in designing and synthesizing novel graphene-based nanomaterials with unique properties and suitable for various applications in the future.
Collapse
Affiliation(s)
- Yuhong Qian
- College of Chemistry, Jilin Normal University, Siping 136000, P. R. China.
| | | | | | | |
Collapse
|
39
|
Wang XS, Yang JM, Ding RJ, Liu XZ, Jiang XB, Yang ZJ, Ling ZM, Hu TX, Wei FX. Fabrication of a Polylactide-Glycolide/Poly-ε-Caprolactone/Dextran/Plastrum Testudinis Extract Composite Anti-Inflammation Nanofiber Membrane via Electrospinning for Annulus Fibrosus Regeneration. J Biomed Nanotechnol 2021; 17:873-888. [PMID: 34082873 DOI: 10.1166/jbn.2021.3070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tissue engineering is a promising approach for the treatment of chronic lower back pain (LBP) caused by intervertebral disc degeneration (IDD) resulting from degeneration and inflammation of annulus fibrosus (AF) tissue. However, scaffold with an anti-inflammatory effect on AF cells has not been reported. In this study, we fabricated a polylactide-glycolide (PLGA)/poly-ε-caprolactone (PCL)Zdextran (DEX) composite membrane loaded with plastrum testudinis extract (PTE), a Traditional Chinese Medicine herbal extract, via electrospinning. The membranes were characterized by mechanical measurements and scanning electron microscopy (SEM). Using an in vitro inflammation model induced by interleukin (IL)-1β, the cytocompatibility and anti-inflammatory effects of the composites were investigated by CCK-8 assay and flow cytometry. Potential regulatory mechanisms were examined by RT-qPCR and Western blotting. The results showed that the P10P8D2 (PLGA 10 g, PCL 8 g, DEX 2 g) composite nanofiber membrane exhibited the most uniform diameter distribution, best mechanical properties, a moderate degradation rate, and the best cytocompatibility characteristics. The optimal concentration of PTE was 120 µg/mL. Importantly, P10P8D2 combined with PTE exhibited anti-inflammatory and cell proliferation promotion effects. Moreover, the NF-κBB/NLRP3/IL-β signaling pathway was inactivated. Our findings suggested that the nanofiber membrane composed of P10P8D2 and PTE has anti-inflammatory and pro-proliferation effects on AF cells. It may provide an effective strategy for AF tissue regeneration.
Collapse
Affiliation(s)
- Xiao-Shuai Wang
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Jia-Ming Yang
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Ren-Jie Ding
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Xi-Zhe Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology Orthopaedic Research Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Xiao-Bing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510080, P. R. China
| | - Zhi-Jian Yang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510030, P. R. China
| | - Ze-Min Ling
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology Orthopaedic Research Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Tian-Xue Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Fu-Xin Wei
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| |
Collapse
|
40
|
Ligorio C, O'Brien M, Hodson NW, Mironov A, Iliut M, Miller AF, Vijayaraghavan A, Hoyland JA, Saiani A. TGF-β3-loaded graphene oxide - self-assembling peptide hybrid hydrogels as functional 3D scaffolds for the regeneration of the nucleus pulposus. Acta Biomater 2021; 127:116-130. [PMID: 33831573 DOI: 10.1016/j.actbio.2021.03.077] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Intervertebral disc (IVD) degeneration is a process that starts in the central nucleus pulposus (NP) and leads to inflammation, extracellular matrix (ECM) degradation, and progressive loss of disc height. Early treatment of IVD degeneration is critical to the reduction of low back pain and related disability. As such, minimally invasive therapeutic approaches that can halt and reverse NP degeneration at the early stages of the disease are needed. Recently, we developed an injectable graphene oxide (GO) - self-assembling peptide FEFKFEFK (F: phenylalanine; K: lysine; E: glutamic acid) hybrid hydrogels as potential delivery platform for cells and/or drugs in the NP. In this current study, we explored the possibility of using the GO present in these hybrid hydrogels as a vehicle for the sequestration and controlled delivery of transforming growth factor beta-3 (TGF-β3), an anabolic growth factor (GF) known to direct NP cell fate and function. For this purpose, we first investigated the potential of GO to bind and sequestrate TGF-β3. We then cultured bovine NP cells in the new functional scaffolds and investigated their response to the presence of GO and TGF-β3. Our results clearly showed that GO flakes can sequestrate TGF-β3 through strong binding interactions resulting in a slow and prolonged release, with the GF remaining active even when bound to the GO flakes. The adsorption of the GF on the GO flakes to create TGF-β3-loaded GO flakes and their subsequent incorporation in the hydrogels through mixing, [(GO/TGF-β3Ads)-F8] hydrogel, led to the upregulation of NP-specific genes, accompanied by the production and deposition of an NP-like ECM, rich in aggrecan and collagen II. NP cells actively interacted with TGF-β3-loaded GO flakes and remodeled the scaffolds through endocytosis. This work highlights the potential of using GO as a nanocarrier for the design of functional hybrid peptide-based hydrogels. STATEMENT OF SIGNIFICANCE: Intervertebral disc (IVD) degeneration is a process that starts in the central nucleus pulposus (NP) and leads to inflammation, extracellular matrix (ECM) degradation, and progressive loss of disc height. As such, minimally invasive therapeutic approaches that can halt and reverse NP degeneration at the early stages of the disease are needed. In this current study, we explored the possibility of using peptide - GO hybrid hydrogels as a vehicle for the sequestration and controlled delivery of transforming growth factor beta-3 (TGF-β3), an anabolic growth factor (GF) known to direct NP cell fate and function.
Collapse
|
41
|
Warren JP, Miles DE, Kapur N, Wilcox RK, Beales PA. Hydrodynamic Mixing Tunes the Stiffness of Proteoglycan-Mimicking Physical Hydrogels. Adv Healthc Mater 2021; 10:e2001998. [PMID: 33943034 PMCID: PMC11468938 DOI: 10.1002/adhm.202001998] [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: 11/16/2020] [Revised: 02/23/2021] [Indexed: 11/09/2022]
Abstract
Self-assembling hydrogels are promising materials for regenerative medicine and tissue engineering. However, designing hydrogels that replicate the 3-4 order of magnitude variation in soft tissue mechanics remains a major challenge. Here hybrid hydrogels are investigated formed from short self-assembling β-fibril peptides, and the glycosaminoglycan chondroitin sulfate (CS), chosen to replicate physical aspects of proteoglycans, specifically natural aggrecan, which provides structural mechanics to soft tissues. Varying the peptide:CS compositional ratio (1:2, 1:10, or 1:20) can tune the mechanics of the gel by one to two orders of magnitude. In addition, it is demonstrated that at any fixed composition, the gel shear modulus can be tuned over approximately two orders of magnitude through varying the initial vortex mixing time. This tuneability arises due to changes in the mesoscale structure of the gel network (fibril width, length, and connectivity), giving rise to both shear-thickening and shear-thinning behavior. The resulting hydrogels range in shear elastic moduli from 0.14 to 220 kPa, mimicking the mechanical variability in a range of soft tissues. The high degree of discrete tuneability of composition and mechanics in these hydrogels makes them particularly promising for matching the chemical and mechanical requirements of different applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- James P. Warren
- School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
- School of Mechanical EngineeringUniversity of LeedsLeedsLS2 9JTUK
- Institute of Medical and Biological EngineeringUniversity of LeedsLeedsLS2 9JTUK
| | - Danielle E. Miles
- School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
- School of Mechanical EngineeringUniversity of LeedsLeedsLS2 9JTUK
- Institute of Medical and Biological EngineeringUniversity of LeedsLeedsLS2 9JTUK
| | - Nikil Kapur
- School of Mechanical EngineeringUniversity of LeedsLeedsLS2 9JTUK
| | - Ruth K. Wilcox
- School of Mechanical EngineeringUniversity of LeedsLeedsLS2 9JTUK
- Institute of Medical and Biological EngineeringUniversity of LeedsLeedsLS2 9JTUK
| | - Paul A. Beales
- School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
- Astbury Centre for Structural BiologyUniversity of LeedsLeedsLS2 9JTUK
- Bragg Centre for Materials ResearchUniversity of LeedsLeedsLS2 9JTUK
| |
Collapse
|
42
|
Neutrally charged self-assembling peptide hydrogel recapitulates in vitro mechanisms of breast cancer progression. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112200. [PMID: 34225853 DOI: 10.1016/j.msec.2021.112200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/05/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022]
Abstract
Self-assembling peptide hydrogels (SAPH) are a popular biomaterial due to their biocompatibility with a wide range of cell types, synthetic design, structural properties that provide a more accurate 3D microenvironment, and potential for cell- and/or drug-delivery system. Mimicking solid tumors in vitro using hydrogels is one method of testing anti-cancer drug efficacy and observing cancerous cell-ECM interactions within a 3D system. In this study, a SAPH, PeptiGel®Alpha1, was used to model in vitro the 3D breast tumor microenvironment. PeptiGel®Alpha1 is composed of entangled nanofibers with consistent diameter and mechanical properties similar to breast cancer that more accurately mimic the stiffness of breast tumor tissue than Matrigel® or collagen type I. PeptiGel®Alpha1 supported the viability and growth of the breast cancer cell lines MCF-7 and MDA-MB-231 and recapitulated key features of solid tumors such as hypoxia and invasion. MCF-7 cells in the hydrogels formed large spheroids resembling acini, while MDA-MB-231 remained dispersed. When treated with tamoxifen, PeptiGel®Alpha1 acted as a barrier, providing drug penetration geometry similar to that in vivo, providing better prediction of the drug effect. Finally, it was observed that MCF-7 cells engulfed the peptide matrix after 14 days, highlighting a potential use in drug delivery. PeptiGel®Alpha1 is a suitable platform for in vitro modeling of breast cancer.
Collapse
|
43
|
Fu K, Wu H, Su Z. Self-assembling peptide-based hydrogels: Fabrication, properties, and applications. Biotechnol Adv 2021; 49:107752. [PMID: 33838284 DOI: 10.1016/j.biotechadv.2021.107752] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/02/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023]
Abstract
The hierarchical formation of self-assembling peptide-based hydrogels (SAPHs) starts from peptide to nanofibers, following with the entanglement into hydrogels with nanofibrous network. Such characteristic structure and extraordinary biocompatibility, and the peptide components endow the SAPHs with diverse applications in biotechnological field. Therefore, the thorough comprehension of SAPHs is significant to broadening their application. In this review, fabrication, properties, and biological applications of the SAPHs are introduced, and the factors influencing the synthesis process as well as the properties of the SAPHs products are also systematically explained. Meanwhile, we conclude the problems to be solved and provide our perspective to the future development of SAPHs in the biotechnology.
Collapse
Affiliation(s)
- Kun Fu
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hanguang Wu
- Beijing Key Laboratory of Clothing Materials R & D and Assessment, Beijing Institute of Fashion Technology, 100029 Beijing, China.
| | - Zhiqiang Su
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Advanced Functional Polymer Composites, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
44
|
Peptide-based scaffolds for the culture and maintenance of primary human hepatocytes. Sci Rep 2021; 11:6772. [PMID: 33762604 PMCID: PMC7990934 DOI: 10.1038/s41598-021-86016-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
We report here the use of a nanofibrous hydrogel as a 3D scaffold for the culture and maintenance of functional primary human hepatocytes. The system is based on the cooperative assembly of a fiber-forming peptide component, fluorenylmethyloxycarbonyl-diphenylalanine (Fmoc-FF), and the integrin-binding functional peptide ligand, Fmoc-arginine-glycine-aspartic acid (Fmoc-RGD) into a nanofibrous gel at physiological pH. This Fmoc-FF/RGD hydrogel was formulated to provide a biomimetic microenvironment with some critical features such as mechanical properties and nanofiber morphology, which were optimized to support hepatocyte culture. The material was shown to support maintenance and function of encapsulated primary human hepatocytes as indicated by actin staining, qRT-PCR, and functional cytochrome P450 assays. The designed gel was shown to outperform Matrigel in cytochrome P450 functional assays. The hydrogel may prove useful for liver development and disease models, as well as providing insights into the design of future implantable scaffolds for the regeneration of liver tissue in patients with liver disease.
Collapse
|
45
|
Carlini AS, Choi W, McCallum NC, Gianneschi NC. pH-Responsive Charge-Conversion Progelator Peptides. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007733. [PMID: 36530181 PMCID: PMC9757809 DOI: 10.1002/adfm.202007733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Indexed: 05/18/2023]
Abstract
A simple strategy for generating stimuli-responsive peptide-based hydrogels via charge-conversion of a self-assembling peptide (SAP) is described. These materials are formulated as soluble, polyanionic peptides, containing maleic acid, citraconic acid, or dimethylmaleic acid masking groups on each lysine residue, which do not form assemblies, but instead flow easily through high gauge needles and catheters. Acid-induced mask hydrolysis renews the zwitterionic nature of the peptides with concomitant and rapid self-assembly via β-sheet formation into rehealable hydrogels. The use of different masks enables one to tune pH responsiveness and assembly kinetics. In anticipation of their potential for in vivo hydrogel delivery and use, progelators exhibit hemocompatibility in whole human blood, and their peptide components are shown to be noncytotoxic. Finally, demonstration of stimuli-induced self-assembly for dye sequestration suggests a simple, non-covalent strategy for small molecule encapsulation in a degradable scaffold. In summary, this simple, scalable masking strategy allows for preparation of responsive, dynamic self-assembling biomaterials. This work sets the stage for implementing biodegradable therapeutic hydrogels that assemble in response to physiological, disease-relevant states of acidosis.
Collapse
Affiliation(s)
- Andrea S Carlini
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Chemistry, International Institute for Nanotechnology, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, USA
| | - Wonmin Choi
- Department of Chemistry, International Institute for Nanotechnology, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, USA
| | - Naneki C McCallum
- Department of Chemistry, International Institute for Nanotechnology, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, USA
| | - Nathan C Gianneschi
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Chemistry, International Institute for Nanotechnology, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, USA
- Department of Materials Science & Engineering, Department of Biomedical Engineering, and Pharmacology, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois 60208, USA
| |
Collapse
|
46
|
Abstract
Intervertebral disc (IVD) degeneration is a leading cause of chronic low back pain (LBP) that results in serious disability and significant economic burden. IVD degeneration alters the disc structure and spine biomechanics, resulting in subsequent structural changes throughout the spine. Currently, treatments of chronic LBP due to IVD degeneration include conservative treatments, such as pain medication and physiotherapy, and surgical treatments, such as removal of herniated disc without or with spinal fusion. However, none of these treatments can completely restore a degenerated disc and its function. Thus, although the exact pathogenesis of disc degeneration remains unclear, there are studies examining the effectiveness of biological approaches, such as growth factor injection, gene therapy, and cell transplantation, in promoting IVD regeneration. Furthermore, tissue engineering using a combination of cell transplantation and biomaterials has emerged as a promising new approach for repair or restoration of degenerated discs. The main purpose of this review was to provide an overview of the current status of tissue engineering applications for IVD regenerative therapy by performing literature searches using PubMed. Significant advances in tissue engineering have opened the door to a new generation of regenerative therapies for the treatment of chronic discogenic LBP.
Collapse
|
47
|
Baumgartner L, Wuertz-Kozak K, Le Maitre CL, Wignall F, Richardson SM, Hoyland J, Ruiz Wills C, González Ballester MA, Neidlin M, Alexopoulos LG, Noailly J. Multiscale Regulation of the Intervertebral Disc: Achievements in Experimental, In Silico, and Regenerative Research. Int J Mol Sci 2021; 22:E703. [PMID: 33445782 PMCID: PMC7828304 DOI: 10.3390/ijms22020703] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a major risk factor of low back pain. It is defined by a progressive loss of the IVD structure and functionality, leading to severe impairments with restricted treatment options due to the highly demanding mechanical exposure of the IVD. Degenerative changes in the IVD usually increase with age but at an accelerated rate in some individuals. To understand the initiation and progression of this disease, it is crucial to identify key top-down and bottom-up regulations' processes, across the cell, tissue, and organ levels, in health and disease. Owing to unremitting investigation of experimental research, the comprehension of detailed cell signaling pathways and their effect on matrix turnover significantly rose. Likewise, in silico research substantially contributed to a holistic understanding of spatiotemporal effects and complex, multifactorial interactions within the IVD. Together with important achievements in the research of biomaterials, manifold promising approaches for regenerative treatment options were presented over the last years. This review provides an integrative analysis of the current knowledge about (1) the multiscale function and regulation of the IVD in health and disease, (2) the possible regenerative strategies, and (3) the in silico models that shall eventually support the development of advanced therapies.
Collapse
Affiliation(s)
- Laura Baumgartner
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY 14623, USA;
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), 81547 Munich, Germany
| | - Christine L. Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK;
| | - Francis Wignall
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Stephen M. Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Judith Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Carlos Ruiz Wills
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Miguel A. González Ballester
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
- Catalan Institution for Research and Advanced Studies (ICREA), Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Jérôme Noailly
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| |
Collapse
|
48
|
Abstract
Supramolecular biopolymers (SBPs) are those polymeric units derived from macromolecules that can assemble with each other by noncovalent interactions. Macromolecular structures are commonly found in living systems such as proteins, DNA/RNA, and polysaccharides. Bioorganic chemistry allows the generation of sequence-specific supramolecular units like SBPs that can be tailored for novel applications in tissue engineering (TE). SBPs hold advantages over other conventional polymers previously used for TE; these materials can be easily functionalized; they are self-healing, biodegradable, stimuli-responsive, and nonimmunogenic. These characteristics are vital for the further development of current trends in TE, such as the use of pluripotent cells for organoid generation, cell-free scaffolds for tissue regeneration, patient-derived organ models, and controlled delivery systems of small molecules. In this review, we will analyse the 3 subtypes of SBPs: peptide-, nucleic acid-, and oligosaccharide-derived. Then, we will discuss the role that SBPs will be playing in TE as dynamic scaffolds, therapeutic scaffolds, and bioinks. Finally, we will describe possible outlooks of SBPs for TE.
Collapse
|
49
|
Imere A, Ligorio C, O'Brien M, Wong JKF, Domingos M, Cartmell SH. Engineering a cell-hydrogel-fibre composite to mimic the structure and function of the tendon synovial sheath. Acta Biomater 2021; 119:140-154. [PMID: 33189954 DOI: 10.1016/j.actbio.2020.11.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 12/27/2022]
Abstract
The repair of tendon injuries is often compromised by post-operative peritendinous adhesions. Placing a physical barrier at the interface between the tendon and the surrounding tissue could potentially solve this problem by reducing adhesion formation. At present, no such system is available for routine use in clinical practice. Here, we propose the development of a bilayer membrane combining a nanofibrous poly(ε-caprolactone) (PCL) electrospun mesh with a layer of self-assembling peptide hydrogel (SAPH) laden with type-B synoviocytes. This bilayer membrane would act as an anti-adhesion system capable of restoring tendon lubrication, while assisting with synovial sheath regeneration. The PCL mesh showed adequate mechanical properties (Young's modulus=19±4 MPa, ultimate tensile stress=9.6±1.7 MPa, failure load=0.5±0.1 N), indicating that the membrane is easy to handle and capable to withstand the frictional forces generated on the tendon's surface during movement (~0.3 N). Morphological analysis confirmed the generation of a mesh with nanosized PCL fibres and small pores (< 3 μm), which prevented fibroblast infiltration to impede extrinsic healing but still allowing diffusion of nutrients and waste. Rheological tests showed that incorporation of SAPH layer allows good lubrication properties when the membrane is articulated against porcine tendon or hypodermis, suggesting that restoration of tendon gliding is possible upon implantation. Moreover, viability and metabolic activity tests indicated that the SAPH was conducive to rabbit synoviocyte growth and proliferation over 28 days of 3D culture, sustaining cell production of specific matrix components, particularly hyaluronic acid. Synoviocyte-laden peptide hydrogel promoted a sustained endogenous production of hyaluronic acid, providing an anti-friction layer that potentially restores the tendon gliding environment.
Collapse
Affiliation(s)
- Angela Imere
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK.; The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK
| | - Cosimo Ligorio
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK.; Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester, UK
| | - Marie O'Brien
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK.; The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK
| | - Jason K F Wong
- Blond McIndoe Laboratories, Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.; Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Marco Domingos
- The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK.; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Sarah H Cartmell
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK.; The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK..
| |
Collapse
|
50
|
Okuno N, Otsuki S, Aoyama J, Nakagawa K, Murakami T, Ikeda K, Hirose Y, Wakama H, Okayoshi T, Okamoto Y, Hirano Y, Neo M. Feasibility of a self-assembling peptide hydrogel scaffold for meniscal defect: An in vivo study in a rabbit model. J Orthop Res 2021; 39:165-176. [PMID: 32852842 DOI: 10.1002/jor.24841] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/19/2020] [Accepted: 08/18/2020] [Indexed: 02/04/2023]
Abstract
The inner avascular zone of the meniscus has limited healing capacity as the area is poorly vascularized. Although peptide hydrogels have been reported to regenerate bone and cartilage, their effect on meniscus regeneration remains unknown. We tested whether the self-assembling peptide hydrogel scaffold KI24RGDS stays in the meniscal lesion and facilitates meniscal repair and regeneration in an induced rabbit meniscal defect model. Full-thickness (2.0 mm diameter) cylindrical defects were introduced into the inner avascular zones of the anterior portions of the medial menisci of rabbit knees (n = 40). Right knee defects were left empty (control group) while the left knee defects were transplanted with peptide hydrogel (KI24RGDS group). Macroscopic meniscus scores were significantly higher in the KI24RGDS group than in the control group at 2, 4, and 8 weeks after surgery. Histological examinations including quantitative and qualitative scores indicated that compared with the control group, the reparative tissue in the meniscus was significantly enhanced in the KI24RGDS group at 2, 4, 8, and 12 weeks after surgery. Immunohistochemical staining showed that the reparative tissue induced by KI24RGDS at 12 weeks postimplantation was positive for Type I and II collagen. KI24RGDS is highly biocompatible and biodegradable, with strong stiffness, and a three dimensional structure mimicking native extracellular matrix and RGDS sequences that enhance cell adhesion and proliferation. This in vivo study demonstrated that KI24RGDS remained in the meniscal lesion and facilitated the repair and regeneration in a rabbit meniscal defect model.
Collapse
Affiliation(s)
- Nobuhiro Okuno
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Shuhei Otsuki
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Jo Aoyama
- Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials, and Bioengineering, Kansai University, Osaka, Japan
| | - Kosuke Nakagawa
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Tomohiko Murakami
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Kuniaki Ikeda
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | | | - Hitoshi Wakama
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Tomohiro Okayoshi
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Yoshinori Okamoto
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Yoshiaki Hirano
- Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials, and Bioengineering, Kansai University, Osaka, Japan
| | - Masashi Neo
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| |
Collapse
|