101
|
Allur Subramanian S, Oh S, Mariadoss AVA, Chae S, Dhandapani S, Parasuraman PS, Song SY, Woo C, Dong X, Choi JY, Kim SJ. Tunable mechanical properties of Mo 3Se 3-poly vinyl alcohol-based/silk fibroin-based nanowire ensure the regeneration mechanism in tenocytes derived from human bone marrow stem cells. Int J Biol Macromol 2022; 210:196-207. [PMID: 35513108 DOI: 10.1016/j.ijbiomac.2022.04.211] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023]
Abstract
Silk fibroin (SF) and poly vinyl alcohol (PVA)-based nanomaterial has exceptional attention in regenerative medicine. However, the preparation of SF and PVA-based nanomaterials in the desired form is complex due to their poor mechanical strength, brittleness, and compatibility. To this end, Mo3Se3 is chosen as a bio-nanowire to fabricate by combining PVA and SF to improve the mechanical properties. Physicochemical and structural features of the Mo3Se3-PVA-SF nanowire hydrogel (Mo3Se3-PVA-SF-NWH) were characterized by field emission scanning electron microscope (FE-SEM). Mechanical properties, degradation ratio, hydrophilicity, water uptake capacity, biocompatibility, and biological activity of the hydrogel were also studied. Superior interactions were formed between the reinforcing molecules of Mo3Se3 and PVA/SF in the hydrogel network by introducing Mo3Se3 nanowire (NW) into the hydrogel. Conversely, Mo3Se3 NW imparts mechanical stability and robustness to the blends (hydrogel) with predictable long-term degradation characteristics. It was proven by in vitro biodegradable rate, and swelling behaviour was varied depending on the concentration of Mo3Se3 NW. Mo3Se3 reinforced the hydrogels and found high porosity with superior biocompatibility. Excellent cellular adaptation was analyzed by MTT assay, live/dead staining, western blot, and quantitative real-time polymerase chain reaction (qRT-PCR). It revealed moderate toxicity at a concentration of 0.02% among the control samples. There was no discernible difference in 0.01% and 0.005% of Mo3Se3-PVA-SF-NWH in tenocytes derived from human bone marrow mesenchymal stem cells (hBMSC). Hence, this Mo3Se3-PVA-SF-NWH might be considered biocompatible due to its biological activities and appropriate mechanical properties. Overall, the Mo3Se3-PVA-SF-NWH might be considered a biocompatible scaffold for the possible biomedical applications of tendon tissue engineering.
Collapse
Affiliation(s)
- Sivakumar Allur Subramanian
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Seungbae Oh
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Arokia Vijaya Anand Mariadoss
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Sudong Chae
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sanjeevram Dhandapani
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Perumalswamy Sekar Parasuraman
- Department of Environmental and Biotechnology, Hallym University, 1 Hallymdeahak-gil, Chuncheon, Gangwon-do 200-702, Republic of Korea
| | - Si Young Song
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Chaeheon Woo
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Xue Dong
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Jae-Young Choi
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea; SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea.
| | - Sung Jae Kim
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea.
| |
Collapse
|
102
|
Application Progress of Modified Chitosan and Its Composite Biomaterials for Bone Tissue Engineering. Int J Mol Sci 2022; 23:ijms23126574. [PMID: 35743019 PMCID: PMC9224397 DOI: 10.3390/ijms23126574] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 12/28/2022] Open
Abstract
In recent years, bone tissue engineering (BTE), as a multidisciplinary field, has shown considerable promise in replacing traditional treatment modalities (i.e., autografts, allografts, and xenografts). Since bone is such a complex and dynamic structure, the construction of bone tissue composite materials has become an attractive strategy to guide bone growth and regeneration. Chitosan and its derivatives have been promising vehicles for BTE owing to their unique physical and chemical properties. With intrinsic physicochemical characteristics and closeness to the extracellular matrix of bones, chitosan-based composite scaffolds have been proved to be a promising candidate for providing successful bone regeneration and defect repair capacity. Advances in chitosan-based scaffolds for BTE have produced efficient and efficacious bio-properties via material structural design and different modifications. Efforts have been put into the modification of chitosan to overcome its limitations, including insolubility in water, faster depolymerization in the body, and blood incompatibility. Herein, we discuss the various modification methods of chitosan that expand its fields of application, which would pave the way for future applied research in biomedical innovation and regenerative medicine.
Collapse
|
103
|
Bercea M. Bioinspired Hydrogels as Platforms for Life-Science Applications: Challenges and Opportunities. Polymers (Basel) 2022; 14:polym14122365. [PMID: 35745941 PMCID: PMC9229923 DOI: 10.3390/polym14122365] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 12/13/2022] Open
Abstract
Hydrogels, as interconnected networks (polymer mesh; physically, chemically, or dynamic crosslinked networks) incorporating a high amount of water, present structural characteristics similar to soft natural tissue. They enable the diffusion of different molecules (ions, drugs, and grow factors) and have the ability to take over the action of external factors. Their nature provides a wide variety of raw materials and inspiration for functional soft matter obtained by complex mechanisms and hierarchical self-assembly. Over the last decade, many studies focused on developing innovative and high-performance materials, with new or improved functions, by mimicking biological structures at different length scales. Hydrogels with natural or synthetic origin can be engineered as bulk materials, micro- or nanoparticles, patches, membranes, supramolecular pathways, bio-inks, etc. The specific features of hydrogels make them suitable for a wide variety of applications, including tissue engineering scaffolds (repair/regeneration), wound healing, drug delivery carriers, bio-inks, soft robotics, sensors, actuators, catalysis, food safety, and hygiene products. This review is focused on recent advances in the field of bioinspired hydrogels that can serve as platforms for life-science applications. A brief outlook on the actual trends and future directions is also presented.
Collapse
Affiliation(s)
- Maria Bercea
- "Petru Poni" Institute of Macromolecular Chemistry, 700487 Iasi, Romania
| |
Collapse
|
104
|
Emerging Polymer Materials in Trackable Endovascular Embolization and Cell Delivery: From Hype to Hope. Biomimetics (Basel) 2022; 7:biomimetics7020077. [PMID: 35735593 PMCID: PMC9221114 DOI: 10.3390/biomimetics7020077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Minimally invasive endovascular embolization is a widely used clinical technique used for the occlusion of blood vessels to treat various diseases. Different occlusive agents ranging from gelatin foam to synthetic polymers such as poly(vinyl alcohol) (PVA) have been commercially used for embolization. However, these agents have some drawbacks, such as undesired toxicity and unintended and uncontrolled occlusion. To overcome these issues, several polymer-based embolic systems are under investigation including biocompatible and biodegradable microspheres, gelling liquid embolic with controlled occlusive features, and trackable microspheres with enhanced safety profiles. This review aims to summarize recent advances in current and emerging polymeric materials as embolization agents with varying material architectures. Furthermore, this review also explores the potential of combining injectable embolic agents and cell therapy to achieve more effective embolization with the promise of outstanding results in treating various devastating diseases. Finally, limitations and challenges in developing next-generation multifunctional embolic agents are discussed to promote advancement in this emerging field.
Collapse
|
105
|
Zhao J, Peng YY, Wang J, Diaz-Dussan D, Tian W, Duan W, Kong L, Hao X, Narain R. Temperature-Responsive Aldehyde Hydrogels with Injectable, Self-Healing, and Tunable Mechanical Properties. Biomacromolecules 2022; 23:2552-2561. [PMID: 35608162 DOI: 10.1021/acs.biomac.2c00260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Injectable and self-healing hydrogels with exemplary biocompatibility and tunable mechanical properties are urgently needed due to their significant advantages for tissue engineering applications. Here, we report a new temperature-responsive aldehyde hydrogel with dual physical-cross-linked networks and injectable and self-healing properties prepared from an ABA-type triblock copolymer, poly{[FPMA(4-formylphenyl methacrylate)-co-DEGMA[di(ethylene glycol) methyl ether methacrylate]-b-MPC(2-methacryloyloxyethyl phosphorylcholine)-b-(FPMA-co-DEGMA)}. The thermoresponsive poly(DEGMA) segments drive the dehydration and hydrophobic interaction, enabling polymer chain winding as the first cross-linking network, when the temperature is raised above the critical gelation temperature. Meanwhile, the benzaldehyde groups offer physical interactions, including hydrogen bonding and hydrophobic and π-π stacking interactions as the second cross-linking network. When increasing the benzaldehyde content in the triblock copolymers from 0 to 8.2 mol %, the critical gelation temperature of the resulted hydrogels dropped from 35.5 to 19.9 °C and the mechanical modulus increased from 21 to 1411 Pa. Owing to the physical-cross-linked networks, the hydrogel demonstrated excellent injectability and self-healing properties. The cell viabilities tested from MTT assays toward both normal lung fibroblast cells (MRC-5) and cancerous cervical (HeLa) cells were found to be 100 and 101%, respectively, for varying polymer concentrations up to 1 mg/mL. The 3D cell encapsulation of the hydrogels was evaluated by a cytotoxicity Live/Dead assay, showing 92% cell viability. With these attractive physiochemical and biological properties, this temperature-responsive aldehyde hydrogel can be a promising candidate as a cell scaffold for tissue engineering.
Collapse
Affiliation(s)
- Jianyang Zhao
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria 3216, Australia.,Manufacturing, CSIRO, Research Way, Clayton, Victoria 3168, Australia.,School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering, University of Alberta, 116 Street and 85th Avenue, Edmonton, Alberta T6G 2G6, Canada
| | - Jinquan Wang
- Guangdong Province Key Laboratory of Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, University of Alberta, 116 Street and 85th Avenue, Edmonton, Alberta T6G 2G6, Canada
| | - Wendy Tian
- Manufacturing, CSIRO, Research Way, Clayton, Victoria 3168, Australia
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Lingxue Kong
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Xiaojuan Hao
- Manufacturing, CSIRO, Research Way, Clayton, Victoria 3168, Australia
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, 116 Street and 85th Avenue, Edmonton, Alberta T6G 2G6, Canada
| |
Collapse
|
106
|
Alshaikh RA, Waeber C, Ryan KB. Polymer based sustained drug delivery to the ocular posterior segment: barriers and future opportunities for the treatment of neovascular pathologies. Adv Drug Deliv Rev 2022; 187:114342. [PMID: 35569559 DOI: 10.1016/j.addr.2022.114342] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
There is an increasing momentum in research and pharmaceutical industry communities to design sustained, non-invasive delivery systems to treat chronic neovascular ocular diseases that affect the posterior segment of the eye including age-related macular degeneration and diabetic retinopathy. Current treatments include VEGF blockers, which have revolutionized the standard of care for patients, but their maximum therapeutic benefit is hampered by the need for recurrent and invasive administration procedures. Currently approved delivery systems intended to address these limitations exploit polymer technology to regulate drug release in a sustained manner. Here, we critically review sustained drug delivery approaches for the treatment of chronic neovascular diseases affecting the ocular posterior segment, with a special emphasis on novel and polymeric technologies spanning the spectrum of preclinical and clinical investigation, and those approved for treatment. The mechanism by which each formulation imparts sustained release, the impact of formulation characteristics on release and foreign body reaction, and special considerations related to the translation of these systems are discussed.
Collapse
Affiliation(s)
| | - Christian Waeber
- School of Pharmacy, University College Cork, Cork, Ireland; Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland; SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland.
| |
Collapse
|
107
|
Damonte G, Maddalena L, Fina A, Cavallo D, Müller AJ, Caputo MR, Mariani A, Monticelli O. On novel hydrogels based on poly(2-hydroxyethyl acrylate) and polycaprolactone with improved mechanical properties prepared by frontal polymerization. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
108
|
Murphy CA, Lim KS, Woodfield TBF. Next Evolution in Organ-Scale Biofabrication: Bioresin Design for Rapid High-Resolution Vat Polymerization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107759. [PMID: 35128736 DOI: 10.1002/adma.202107759] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/30/2022] [Indexed: 06/14/2023]
Abstract
The field of bioprinting has made significant advancements in recent years and allowed for the precise deposition of biomaterials and cells. However, within this field lies a major challenge, which is developing high resolution constructs, with complex architectures. In an effort to overcome these challenges a biofabrication technique known as vat polymerization is being increasingly investigated due to its high fabrication accuracy and control of resolution (µm scale). Despite the progress made in developing hydrogel precursors for bioprinting techniques, such as extrusion-based bioprinting, there is a major lack in developing hydrogel precursor bioresins for vat polymerization. This is due to the specific unique properties and characteristics required for vat polymerization, from lithography to the latest volumetric printing. This is of major concern as the shortage of bioresins available has a significant impact on progressing this technology and exploring its full potential, including speed, resolution, and scale. Therefore, this review discusses the key requirements that need to be addressed in successfully developing a bioresin. The influence of monomer architecture and bioresin composition on printability is described, along with key fundamental parameters that can be altered to increase printing accuracy. Finally, recent advancements in bioresins are discussed together with future directions.
Collapse
Affiliation(s)
- Caroline A Murphy
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering and Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering and Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering and Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering and Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
| |
Collapse
|
109
|
Hakim M, Kermanshah L, Abouali H, Hashemi HM, Yari A, Khorasheh F, Alemzadeh I, Vossoughi M. Unraveling Cancer Metastatic Cascade Using Microfluidics-based Technologies. Biophys Rev 2022; 14:517-543. [PMID: 35528034 PMCID: PMC9043145 DOI: 10.1007/s12551-022-00944-8] [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: 07/15/2021] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer has long been a leading cause of death. The primary tumor, however, is not the main cause of death in more than 90% of cases. It is the complex process of metastasis that makes cancer deadly. The invasion metastasis cascade is the multi-step biological process of cancer cell dissemination to distant organ sites and adaptation to the new microenvironment site. Unraveling the metastasis process can provide great insight into cancer death prevention or even treatment. Microfluidics is a promising platform, that provides a wide range of applications in metastasis-related investigations. Cell culture microfluidic technologies for in vitro modeling of cancer tissues with fluid flow and the presence of mechanical factors have led to the organ-on-a-chip platforms. Moreover, microfluidic systems have also been exploited for capturing and characterization of circulating tumor cells (CTCs) that provide crucial information on the metastatic behavior of a tumor. We present a comprehensive review of the recent developments in the application of microfluidics-based systems for analysis and understanding of the metastasis cascade from a wider perspective.
Collapse
Affiliation(s)
- Maziar Hakim
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Leyla Kermanshah
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hesam Abouali
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hanieh Mohammad Hashemi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Alireza Yari
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Farhad Khorasheh
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Iran Alemzadeh
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Manouchehr Vossoughi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
110
|
Dibazar ZE, Mohammadpour M, Samadian H, Zare S, Azizi M, Hamidi M, Elboutachfaiti R, Petit E, Delattre C. Bacterial Polyglucuronic Acid/Alginate/Carbon Nanofibers Hydrogel Nanocomposite as a Potential Scaffold for Bone Tissue Engineering. MATERIALS 2022; 15:ma15072494. [PMID: 35407826 PMCID: PMC8999617 DOI: 10.3390/ma15072494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
3D nanocomposite scaffolds have attracted significant attention in bone tissue engineering applications. In the current study, we fabricated a 3D nanocomposite scaffold based on a bacterial polyglucuronic acid (PGU) and sodium alginate (Alg) composite with carbon nanofibers (CNFs) as the bone tissue engineering scaffold. The CNFs were obtained from electrospun polyacrylonitrile nanofibers through heat treatment. The fabricated CNFs were incorporated into a PGU/Alg polymeric solution, which was physically cross-linked using CaCl2 solution. The fabricated nanocomposites were characterized to evaluate the internal structure, porosity, swelling kinetics, hemocompatibility, and cytocompatibility. The characterizations indicated that the nanocomposites have a porous structure with interconnected pores architecture, proper water absorption, and retention characteristics. The in vitro studies revealed that the nanocomposites were hemocompatible with negligible hemolysis induction. The cell viability assessment showed that the nanocomposites were biocompatible and supported bone cell growth. These results indicated that the fabricated bacterial PGU/Alg/CNFs hydrogel nanocomposite exhibited appropriate properties and can be considered a new biomaterial for bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Zahra Ebrahimvand Dibazar
- Department of Oral and Maxillo Facial Medicine, Faculty of Dentistry, Tabriz Asad University of Medical Sciences, Tabriz 5166616471, Iran;
| | - Mahnaz Mohammadpour
- Department of Chemistry, Faculty of Sciences, Tarbiat Modares University, Tehran 1411713116, Iran;
| | - Hadi Samadian
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Correspondence: (H.S.); (R.E.); (C.D.)
| | - Soheila Zare
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan 7797845157, Iran;
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran;
| | - Masoud Hamidi
- BioMatter-Biomass Transformation Lab (BTL), École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, 1050 Brussels, Belgium;
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht 4188794755, Iran
| | - Redouan Elboutachfaiti
- UMRT INRAE 1158 BioEcoAgro, Laboratoire BIOPI, Université de Picardie Jules Verne, IUT d’Amiens, 80025 Amiens, France;
- Correspondence: (H.S.); (R.E.); (C.D.)
| | - Emmanuel Petit
- UMRT INRAE 1158 BioEcoAgro, Laboratoire BIOPI, Université de Picardie Jules Verne, IUT d’Amiens, 80025 Amiens, France;
| | - Cédric Delattre
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, Institut Pascal, 63000 Clermont-Ferrand, France
- Institut Universitaire de France (IUF), 1 Rue Descartes, 75005 Paris, France
- Correspondence: (H.S.); (R.E.); (C.D.)
| |
Collapse
|
111
|
Injectable Hydrogel Based on Protein-Polyester Microporous Network as an Implantable Niche for Active Cell Recruitment. Pharmaceutics 2022; 14:pharmaceutics14040709. [PMID: 35456546 PMCID: PMC9024632 DOI: 10.3390/pharmaceutics14040709] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Despite the potential of hydrogel-based localized cancer therapies, their efficacy can be limited by cancer recurrence. Therefore, it is of great significance to develop a hydrogel system that can provoke robust and durable immune response in the human body. This study has developed an injectable protein-polymer-based porous hydrogel network composed of lysozyme and poly(ε-caprolactone-co-lactide)-b-poly(ethylene glycol)-b-poly(ε-caprolactone-co-lactide (PCLA) (Lys-PCLA) bioconjugate for the active recruitment dendritic cells (DCs). The Lys-PCLA bioconjugates are prepared using thiol-ene reaction between thiolated lysozyme (Lys-SH) and acrylated PCLA (PCLA-Ac). The free-flowing Lys-PCLA bioconjugate sols at low temperature transformed to immovable gel at the physiological condition and exhibited stability upon dilution with buffers. According to the in vitro toxicity test, the Lys-PCLA bioconjugate and PCLA copolymer were non-toxic to RAW 263.7 cells at higher concentrations (1000 µg/mL). In addition, subcutaneous administration of Lys-PCLA bioconjugate sols formed stable hydrogel depot instantly, which suggested the in situ gel forming ability of the bioconjugate. Moreover, the Lys-PCLA bioconjugate hydrogel depot formed at the interface between subcutaneous tissue and dermis layers allowed the active migration and recruitment of DCs. As suggested by these results, the in-situ forming injectable Lys-PCLA bioconjugate hydrogel depot may serve as an implantable immune niche for the recruitment and modification of DCs.
Collapse
|
112
|
Addition of High Acyl Gellan Gum to Low Acyl Gellan Gum Enables the Blends 3D Bioprintable. Gels 2022; 8:gels8040199. [PMID: 35448100 PMCID: PMC9030627 DOI: 10.3390/gels8040199] [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: 01/25/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 12/02/2022] Open
Abstract
Long-term stability of gellan gum (GG) at physiological conditions is expected, as very low concentration of divalent ions are required for crosslinking, as compared to alginate—which is extensively used for tissue engineering (TE) applications. Hence, GG is proposed as an ideal candidate to substitute alginate for TE. Deacylated (low acyl; LA) GG forms brittle gels, thus only low concentrations were used for cell encapsulation, whereas acylated (high acyl; HA) GG forms weak/soft gels. 3D bioprinting using pure LAGG or HAGG is not possible owing to their rheological properties. Here, we report development and characterization of bioprintable blends of LAGG and HAGG. Increase in HAGG in the blends improved shear recovery and shape fidelity of printed scaffolds. Low volumetric swelling observed in cell culture conditions over 14 days indicates stability. Volumetric scaffolds were successfully printed and their mechanical properties were determined by uniaxial compressive testing. Mesenchymal stem cells bioprinted in blends of 3% LAGG and 3% HAGG survived the printing process showing >80% viability; a gradual decrease in cell numbers was observed over 21 days of culture. However, exploiting intrinsic advantages of 3D bioprinting, LAGG/HAGG blends open up numerous possibilities to improve and/or tailor various aspects required for TE.
Collapse
|
113
|
He T, Qiao S, Ma C, Peng Z, Wu Z, Ma C, Han L, Deng Q, Zhang T, Zhu Y, Pan G. FEK self-assembled peptide hydrogels facilitate primary hepatocytes culture and pharmacokinetics screening. J Biomed Mater Res B Appl Biomater 2022; 110:2015-2027. [PMID: 35301798 DOI: 10.1002/jbm.b.35056] [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: 05/12/2021] [Revised: 09/11/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Abstract
A FEFEFKFK (FEK, F, phenylalaninyl; E, glutamyl; K, lysinyl)-based self-assembling peptide hydrogel (FEK-SAPH) was developed to replace sandwich culture (SC) for improved culture of primary hepatocytes in vitro. Under neutral conditions, FEK self-assembles to form β-sheet nanofibers, which in turn form FEK-SAPH. For the culture of rat primary hepatocytes (RPH), the use of FEK-SAPH simplified operation steps and promoted excellent cell-cell interactions while maintaining the SC-related RPH polarity trend. Compared with SC, FEK-SAPH cultured RPH for 14 days, the bile duct network was formed, the secretion of albumin and urea was improved, and the metabolic clearance rate based on cytochrome P450 (CYPs) was comparable. In FEK-SAPH culture, the expression level of the biliary efflux transporter bile salt export pump increased by 230.7%, while the biliary excretion index value of deuterium-labeled sodium taurocholate (d8-TCA) differed slightly from the SC value (72% and 77%, respectively, p = .0195). The inhibitory effect of cholestasis drugs on FEK-SAPH was significantly higher than that of SC. In FEK-SAPH, hepatoprotective drugs were more effective in antagonizing hepatotoxicity induced by lithocholic acid (LCA). FEK-SAPH cultured RPH with hepatoprotective drugs can better recover from LCA-induced damage. In summary, FEK-SAPH can be used as a substitute for SC for pharmacokinetic screening to evaluate the drug absorption, disposition, metabolism, excretion, and toxicity (ADMET) in hepatocytes.
Collapse
Affiliation(s)
- Ting He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shida Qiao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chen Ma
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhaoliang Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhitao Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenhui Ma
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Li Han
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiangqiang Deng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tianwei Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
114
|
Radulescu DM, Neacsu IA, Grumezescu AM, Andronescu E. New Insights of Scaffolds Based on Hydrogels in Tissue Engineering. Polymers (Basel) 2022; 14:799. [PMID: 35215710 PMCID: PMC8875010 DOI: 10.3390/polym14040799] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
In recent years, biomaterials development and characterization for new applications in regenerative medicine or controlled release represent one of the biggest challenges. Tissue engineering is one of the most intensively studied domain where hydrogels are considered optimum applications in the biomedical field. The delicate nature of hydrogels and their low mechanical strength limit their exploitation in tissue engineering. Hence, developing new, stronger, and more stable hydrogels with increased biocompatibility, is essential. However, both natural and synthetic polymers possess many limitations. Hydrogels based on natural polymers offer particularly high biocompatibility and biodegradability, low immunogenicity, excellent cytocompatibility, variable, and controllable solubility. At the same time, they have poor mechanical properties, high production costs, and low reproducibility. Synthetic polymers come to their aid through superior mechanical strength, high reproducibility, reduced costs, and the ability to regulate their composition to improve processes such as hydrolysis or biodegradation over variable periods. The development of hydrogels based on mixtures of synthetic and natural polymers can lead to the optimization of their properties to obtain ideal scaffolds. Also, incorporating different nanoparticles can improve the hydrogel's stability and obtain several biological effects. In this regard, essential oils and drug molecules facilitate the desired biological effect or even produce a synergistic effect. This study's main purpose is to establish the main properties needed to develop sustainable polymeric scaffolds. These scaffolds can be applied in tissue engineering to improve the tissue regeneration process without producing other side effects to the environment.
Collapse
Affiliation(s)
- Denisa-Maria Radulescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
| | - Ionela Andreea Neacsu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
- Academy of Romanian Scientists, 54 Independentei, 050094 Bucharest, Romania
- National Research Center for Micro and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
| | - Alexandru-Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
- Academy of Romanian Scientists, 54 Independentei, 050094 Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
- Academy of Romanian Scientists, 54 Independentei, 050094 Bucharest, Romania
- National Research Center for Micro and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
| |
Collapse
|
115
|
Kurian AG, Singh RK, Patel KD, Lee JH, Kim HW. Multifunctional GelMA platforms with nanomaterials for advanced tissue therapeutics. Bioact Mater 2022; 8:267-295. [PMID: 34541401 PMCID: PMC8424393 DOI: 10.1016/j.bioactmat.2021.06.027] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Polymeric hydrogels are fascinating platforms as 3D scaffolds for tissue repair and delivery systems of therapeutic molecules and cells. Among others, methacrylated gelatin (GelMA) has become a representative hydrogel formulation, finding various biomedical applications. Recent efforts on GelMA-based hydrogels have been devoted to combining them with bioactive and functional nanomaterials, aiming to provide enhanced physicochemical and biological properties to GelMA. The benefits of this approach are multiple: i) reinforcing mechanical properties, ii) modulating viscoelastic property to allow 3D printability of bio-inks, iii) rendering electrical/magnetic property to produce electro-/magneto-active hydrogels for the repair of specific tissues (e.g., muscle, nerve), iv) providing stimuli-responsiveness to actively deliver therapeutic molecules, and v) endowing therapeutic capacity in tissue repair process (e.g., antioxidant effects). The nanomaterial-combined GelMA systems have shown significantly enhanced and extraordinary behaviors in various tissues (bone, skin, cardiac, and nerve) that are rarely observable with GelMA. Here we systematically review these recent efforts in nanomaterials-combined GelMA hydrogels that are considered as next-generation multifunctional platforms for tissue therapeutics. The approaches used in GelMA can also apply to other existing polymeric hydrogel systems.
Collapse
Affiliation(s)
- Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K. Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kapil D. Patel
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, WC1X8LD, UK
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
116
|
Güngör Z, Ozay H. Synthesis of new type temperature and pH sensitive hydrogels using drug-based p-(methacryloyloxy)acetanilide monomer and their usage as controlled drug carrier material. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2022. [DOI: 10.1080/10601325.2022.2028552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Zeynep Güngör
- School of Graduate Studies, Department of Chemistry, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Hava Ozay
- Laboratory of Inorganic Materials, Department of Chemistry, Faculty of Science and Arts, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
117
|
Ozgun A, Lomboni D, Arnott H, Staines WA, Woulfe J, Variola F. Biomaterial-based strategies for in vitro neural models. Biomater Sci 2022; 10:1134-1165. [PMID: 35023513 DOI: 10.1039/d1bm01361k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In vitro models have been used as a complementary tool to animal studies in understanding the nervous system's physiological mechanisms and pathological disorders, while also serving as platforms to evaluate the safety and efficiency of therapeutic candidates. Following recent advances in materials science, micro- and nanofabrication techniques and cell culture systems, in vitro technologies have been rapidly gaining the potential to bridge the gap between animal and clinical studies by providing more sophisticated models that recapitulate key aspects of the structure, biochemistry, biomechanics, and functions of human tissues. This was made possible, in large part, by the development of biomaterials that provide cells with physicochemical features that closely mimic the cellular microenvironment of native tissues. Due to the well-known material-driven cellular response and the importance of mimicking the environment of the target tissue, the selection of optimal biomaterials represents an important early step in the design of biomimetic systems to investigate brain structures and functions. This review provides a comprehensive compendium of commonly used biomaterials as well as the different fabrication techniques employed for the design of neural tissue models. Furthermore, the authors discuss the main parameters that need to be considered to develop functional platforms not only for the study of brain physiological functions and pathological processes but also for drug discovery/development and the optimization of biomaterials for neural tissue engineering.
Collapse
Affiliation(s)
- Alp Ozgun
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - David Lomboni
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Hallie Arnott
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - William A Staines
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - John Woulfe
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,The Ottawa Hospital, Ottawa, Canada
| | - Fabio Variola
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada.,The Ottawa Hospital, Ottawa, Canada.,Children's Hospital of Eastern Ontario (CHEO), Ottawa, Canada
| |
Collapse
|
118
|
Chaudhary S, Chakraborty E. Hydrogel based tissue engineering and its future applications in personalized disease modeling and regenerative therapy. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022; 11:3. [PMID: 35005036 PMCID: PMC8725962 DOI: 10.1186/s43088-021-00172-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Evolution in the in vitro cell culture from conventional 2D to 3D technique has been a significant accomplishment. The 3D culture models have provided a close and better insight into the physiological study of the human body. The increasing demand for organs like liver, kidney, and pancreas for transplantation, rapid anti-cancer drug screening, and the limitations associated with the use of animal models have attracted the interest of researchers to explore 3D organ culture. MAIN BODY Natural, synthetic, and hybrid material-based hydrogels are being used as scaffolds in 3D culture and provide 'close-to-in vivo' structures. Organoids: the stem cell-derived small size 3D culture systems are now favored due to their ability to mimic the in-vivo conditions of organ or tissue and this characteristic has made it eligible for a variety of clinical applications, drug discovery and regenerative medicine are a few of the many areas of application. The use of animal models for clinical applications has been a long-time ethical and biological challenge to get accurate outcomes. 3D bioprinting has resolved the issue of vascularization in organoid culture to a great extent by its layer-by-layer construction approach. The 3D bioprinted organoids have a popular application in personalized disease modeling and rapid drug development and therapeutics. SHORT CONCLUSIONS This review paper, focuses on discussing the novel organoid culture approach, its advantages and limitations, and potential applications in a variety of life science areas namely cancer research, cell therapy, tissue engineering, and personalized medicine and drug discovery. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Shikha Chaudhary
- SRM Institute of Science & Technology, Chennai, Tamil Nadu 603203 India
| | - Eliza Chakraborty
- Medical Translational Biotechnology Lab, Prof of Department of Biotechnology, Head of the Department of DST-Fist Center (Sponsored By Ministry of Science & Technology, Government of India), Meerut Institute of Engineering and Technology (MIET), Meerut, Uttar Pradesh 250002 India
| |
Collapse
|
119
|
Li M, Sun D, Zhang J, Wang Y, Wei Q, Wang Y. Application and development of 3D bioprinting in cartilage tissue engineering. Biomater Sci 2022; 10:5430-5458. [DOI: 10.1039/d2bm00709f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bioprinting technology can build complex tissue structures and has the potential to fabricate engineered cartilage with bionic structures for achieving cartilage defect repair/regeneration.
Collapse
Affiliation(s)
- Mingyang Li
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Daocen Sun
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Juan Zhang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yanmei Wang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Qinghua Wei
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yanen Wang
- Industry Engineering Department, School of Mechanical Engineering, Northwestern Polytechnical University, Xi'an 710072, P.R. China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
120
|
Köhne M, Behrens CS, Stüdemann T, von Bibra C, Querdel E, Shibamiya A, Geertz B, Olfe J, Hüners I, Jockenhövel S, Hübler M, Eschenhagen T, Sachweh JS, Weinberger F, Biermann D. OUP accepted manuscript. Eur J Cardiothorac Surg 2022; 62:6537620. [PMID: 35218664 PMCID: PMC9373941 DOI: 10.1093/ejcts/ezac111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/13/2022] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Maria Köhne
- Department of Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Charlotta Sophie Behrens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Tim Stüdemann
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Constantin von Bibra
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Eva Querdel
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Aya Shibamiya
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Birgit Geertz
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Olfe
- Department of Pediatric Cardiology, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ida Hüners
- Department of Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Jockenhövel
- Department of Biohybrid & Medical Textiles (Biotex), RWTH Aachen University, Aachen, Germany
| | - Michael Hübler
- Department of Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Jörg Siegmar Sachweh
- Department of Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Daniel Biermann
- Department of Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Corresponding author. Department of Congenital and Pediatric Heart Surgery, Children's Heart Clinic, University Heart & Vascular Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany. Tel: +49-40-7410-58221; e-mail: (D. Biermann)
| |
Collapse
|
121
|
You R, Shi L, Chen S, Liu Q, Zhang L, Yin L, Yang R, Guan YQ. Fabrication of Resveratrol-Loaded Scaffolds and Their Application for Delaying Cell Senescence In Vitro. Macromol Biosci 2021; 22:e2100440. [PMID: 34919323 DOI: 10.1002/mabi.202100440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/10/2021] [Indexed: 11/08/2022]
Abstract
In this research, resveratrol (RSV)-loaded scaffolds have been prepared to control the release of resveratrol and used to delay hepatic stellate cell (HSC) senescence in vitro. The functional carboxyl group-COOH is first introduced to the surface of poly(ε-caprolactone/d,l-lactide) (P(CL-DLLA)) under the coadministration of ultra-violet (UV) treatment and photo initiator and then resveratrol are conjugated onto the surface of the modified scaffolds through esterification. The characterization of the structure of RSV-AA-P(CL-DLLA) shows that resveratrol has been successfully conjugated onto the modified surface. Cell growth exhibits a higher level of cell viability and much more obvious agglomeration on the surface of the synthetic RSV-AA-P(CL-DLLA). Meanwhile the activity of senescence-associated β-galactosidase (SA-β-gal) and reactive oxygen species (ROS) is downgulated for cells on RSV-AA-P(CL-DLLA), which suggests that cell senescence is delayed on RSV-AA-P(CL-DLLA). And then it is attested that cells have a lower level of p53 but SIRT1 expression is upregulated on RSV-AA-P(CL-DLLA), which might be related to resveratrol release from RSV-AA-P(CL-DLLA). It also suggested cell senescence on RSV-AA-P(CL-DLLA) has been regulated by p53 and the SIRT1 signaling pathway. In all, the present study shows that RSV-AA-P(CL-DLLA) can be successfully prepared to promote cell growth and delay cell senescence and could be used for cell-based therapy in tissue engineering.
Collapse
Affiliation(s)
- Rong You
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Lu Shi
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Surong Chen
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Qingpeng Liu
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Lingkun Zhang
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Runcai Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou, 510631, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
122
|
Muntz I, Fenu M, van Osch GJVM, Koenderink G. The role of cell-matrix interactions in connective tissue mechanics. Phys Biol 2021; 19. [PMID: 34902848 DOI: 10.1088/1478-3975/ac42b8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/13/2021] [Indexed: 11/12/2022]
Abstract
Living tissue is able to withstand large stresses in everyday life, yet it also actively adapts to dynamic loads. This remarkable mechanical behaviour emerges from the interplay between living cells and their non-living extracellular environment. Here we review recent insights into the biophysical mechanisms involved in the reciprocal interplay between cells and the extracellular matrix and how this interplay determines tissue mechanics, with a focus on connective tissues. We first describe the roles of the main macromolecular components of the extracellular matrix in regards to tissue mechanics. We then proceed to highlight the main routes via which cells sense and respond to their biochemical and mechanical extracellular environment. Next we introduce the three main routes via which cells can modify their extracellular environment: exertion of contractile forces, secretion and deposition of matrix components, and matrix degradation. Finally we discuss how recent insights in the mechanobiology of cell-matrix interactions are furthering our understanding of the pathophysiology of connective tissue diseases and cancer, and facilitating the design of novel strategies for tissue engineering.
Collapse
Affiliation(s)
- Iain Muntz
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HC, NETHERLANDS
| | - Michele Fenu
- Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gerjo J V M van Osch
- Orthopaedics; Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gijsje Koenderink
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HZ, NETHERLANDS
| |
Collapse
|
123
|
Khan F, Atif M, Haseen M, Kamal S, Khan MS, Shahid S, Nami SAA. Synthesis, classification and properties of hydrogels: their applications in drug delivery and agriculture. J Mater Chem B 2021; 10:170-203. [PMID: 34889937 DOI: 10.1039/d1tb01345a] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Absorbent polymers or hydrogel polymer materials have an enhanced water retention capacity and are widely used in agriculture and medicine. The controlled release of bioactive molecules (especially drug proteins) by hydrogels and the encapsulation of living cells are some of the active areas of drug discovery research. Hydrogel-based delivery systems may result in a therapeutically advantageous outcome for drug delivery. They can provide various sequential therapeutic agents including macromolecular drugs, small molecule drugs, and cells to control the release of molecules. Due to their controllable degradability, ability to protect unstable drugs from degradation and flexible physical properties, hydrogels can be used as a platform in which various chemical and physical interactions with encapsulated drugs for controlled release in the system can be studied. Practically, hydrogels that possess biodegradable properties have aroused greater interest in drug delivery systems. The original three-dimensional structure gets broken down into non-toxic substances, thus confirming the excellent biocompatibility of the gel. Chemical crosslinking is a resource-rich method for forming hydrogels with excellent mechanical strength. But in some cases the crosslinker used in the synthesis of the hydrogels may cause some toxicity. However, the physically cross-linked hydrogel preparative method is an alternative solution to overcome the toxicity of cross-linkers. Hydrogels that are responsive to stimuli formed from various natural and synthetic polymers can show significant changes in their properties under external stimuli such as temperature, pH, light, ion changes, and redox potential. Stimulus-responsive hydrogels have a wider range of applications in biomedicine including drug delivery, gene delivery and tissue regeneration. Stimulus-responsive hydrogels loaded with multiple drugs show controlled and sustained drug release and can act as drug carriers. By integrating stimulus-responsive hydrogels, such as those with improved thermal responsiveness, pH responsiveness and dual responsiveness, into textile materials, advanced functions can be imparted to the textile materials, thereby improving the moisture and water retention performance, environmental responsiveness, aesthetic appeal, display and comfort of textiles. This review explores the stimuli-responsive hydrogels in drug delivery systems and examines super adsorbent hydrogels and their application in the field of agriculture.
Collapse
Affiliation(s)
- Faisal Khan
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohd Atif
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohd Haseen
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Shahid Kamal
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohd Shoeb Khan
- Interdisciplinary Nanotechnology Centre, Aligarh Muslim University, Aligarh 202002, India
| | - Shumaila Shahid
- Division of Plant Pathology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Shahab A A Nami
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
124
|
Das S, Das D. Rational Design of Peptide-based Smart Hydrogels for Therapeutic Applications. Front Chem 2021; 9:770102. [PMID: 34869218 PMCID: PMC8635208 DOI: 10.3389/fchem.2021.770102] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Peptide-based hydrogels have captivated remarkable attention in recent times and serve as an excellent platform for biomedical applications owing to the impressive amalgamation of unique properties such as biocompatibility, biodegradability, easily tunable hydrophilicity/hydrophobicity, modular incorporation of stimuli sensitivity and other functionalities, adjustable mechanical stiffness/rigidity and close mimicry to biological molecules. Putting all these on the same plate offers smart soft materials that can be used for tissue engineering, drug delivery, 3D bioprinting, wound healing to name a few. A plethora of work has been accomplished and a significant progress has been realized using these peptide-based platforms. However, designing hydrogelators with the desired functionalities and their self-assembled nanostructures is still highly serendipitous in nature and thus a roadmap providing guidelines toward designing and preparing these soft-materials and applying them for a desired goal is a pressing need of the hour. This review aims to provide a concise outline for that purpose and the design principles of peptide-based hydrogels along with their potential for biomedical applications are discussed with the help of selected recent reports.
Collapse
Affiliation(s)
- Saurav Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| | - Debapratim Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
125
|
Mendoza-Martinez AK, Loessner D, Mata A, Azevedo HS. Modeling the Tumor Microenvironment of Ovarian Cancer: The Application of Self-Assembling Biomaterials. Cancers (Basel) 2021; 13:5745. [PMID: 34830897 PMCID: PMC8616551 DOI: 10.3390/cancers13225745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OvCa) is one of the leading causes of gynecologic malignancies. Despite treatment with surgery and chemotherapy, OvCa disseminates and recurs frequently, reducing the survival rate for patients. There is an urgent need to develop more effective treatment options for women diagnosed with OvCa. The tumor microenvironment (TME) is a key driver of disease progression, metastasis and resistance to treatment. For this reason, 3D models have been designed to represent this specific niche and allow more realistic cell behaviors compared to conventional 2D approaches. In particular, self-assembling peptides represent a promising biomaterial platform to study tumor biology. They form nanofiber networks that resemble the architecture of the extracellular matrix and can be designed to display mechanical properties and biochemical motifs representative of the TME. In this review, we highlight the properties and benefits of emerging 3D platforms used to model the ovarian TME. We also outline the challenges associated with using these 3D systems and provide suggestions for future studies and developments. We conclude that our understanding of OvCa and advances in materials science will progress the engineering of novel 3D approaches, which will enable the development of more effective therapies.
Collapse
Affiliation(s)
- Ana Karen Mendoza-Martinez
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Daniela Loessner
- Department of Chemical Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia;
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., 01069 Dresden, Germany
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
126
|
Zennifer A, Manivannan S, Sethuraman S, Kumbar SG, Sundaramurthi D. 3D bioprinting and photocrosslinking: emerging strategies & future perspectives. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112576. [DOI: 10.1016/j.msec.2021.112576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022]
|
127
|
Dogan E, Kisim A, Bati-Ayaz G, Kubicek GJ, Pesen-Okvur D, Miri AK. Cancer Stem Cells in Tumor Modeling: Challenges and Future Directions. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100017. [PMID: 34927168 PMCID: PMC8680587 DOI: 10.1002/anbr.202100017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Microfluidic tumors-on-chips models have revolutionized anticancer therapeutic research by creating an ideal microenvironment for cancer cells. The tumor microenvironment (TME) includes various cell types and cancer stem cells (CSCs), which are postulated to regulate the growth, invasion, and migratory behavior of tumor cells. In this review, the biological niches of the TME and cancer cell behavior focusing on the behavior of CSCs are summarized. Conventional cancer models such as three-dimensional cultures and organoid models are reviewed. Opportunities for the incorporation of CSCs with tumors-on-chips are then discussed for creating tumor invasion models. Such models will represent a paradigm shift in the cancer community by allowing oncologists and clinicians to predict better which cancer patients will benefit from chemotherapy treatments.
Collapse
Affiliation(s)
- Elvan Dogan
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028
| | - Asli Kisim
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey
| | - Gizem Bati-Ayaz
- Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Gregory J. Kubicek
- Department of Radiation Oncology, MD Anderson Cancer Center at Cooper, 2 Cooper Plaza, Camden, NJ 08103
| | - Devrim Pesen-Okvur
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey; Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028; School of Medical Engineering, Science, and Health, Rowan University, Camden, NJ 08103
| |
Collapse
|
128
|
Devi V. K. A, Shyam R, Palaniappan A, Jaiswal AK, Oh TH, Nathanael AJ. Self-Healing Hydrogels: Preparation, Mechanism and Advancement in Biomedical Applications. Polymers (Basel) 2021; 13:3782. [PMID: 34771338 PMCID: PMC8587783 DOI: 10.3390/polym13213782] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
Polymeric hydrogels are widely explored materials for biomedical applications. However, they have inherent limitations like poor resistance to stimuli and low mechanical strength. This drawback of hydrogels gave rise to ''smart self-healing hydrogels'' which autonomously repair themselves when ruptured or traumatized. It is superior in terms of durability and stability due to its capacity to reform its shape, injectability, and stretchability thereby regaining back the original mechanical property. This review focuses on various self-healing mechanisms (covalent and non-covalent interactions) of these hydrogels, methods used to evaluate their self-healing properties, and their applications in wound healing, drug delivery, cell encapsulation, and tissue engineering systems. Furthermore, composite materials are used to enhance the hydrogel's mechanical properties. Hence, findings of research with various composite materials are briefly discussed in order to emphasize the healing capacity of such hydrogels. Additionally, various methods to evaluate the self-healing properties of hydrogels and their recent advancements towards 3D bioprinting are also reviewed. The review is concluded by proposing several pertinent challenges encountered at present as well as some prominent future perspectives.
Collapse
Affiliation(s)
- Anupama Devi V. K.
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (A.D.V.K.); (R.S.); (A.P.)
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Rohin Shyam
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (A.D.V.K.); (R.S.); (A.P.)
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Arunkumar Palaniappan
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (A.D.V.K.); (R.S.); (A.P.)
| | - Amit Kumar Jaiswal
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (A.D.V.K.); (R.S.); (A.P.)
| | - Tae-Hwan Oh
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea;
| | - Arputharaj Joseph Nathanael
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (A.D.V.K.); (R.S.); (A.P.)
| |
Collapse
|
129
|
Orr A, Wilson P, Stotesbury T. Calcium-Alginate Tissue Gels (CATG): Proof-of-concept biomaterial development. Forensic Sci Int 2021; 329:111055. [PMID: 34688973 DOI: 10.1016/j.forsciint.2021.111055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/22/2021] [Accepted: 10/09/2021] [Indexed: 11/29/2022]
Abstract
Hydrogels are desirable materials to the field of forensic science and offer many advantages for use as tissue simulants in research and training scenarios. In this work, we demonstrate a proof-of-concept study for our biomaterial described as the Calcium-Alginate Tissue Gel (CATG). CATG biomaterials integrate functional DNA strands designed to amplify with known human primer sets for genetic profiling. Our range of CATG materials demonstrate successful DNA extraction, PCR amplification and genotyping when both fresh and aged for 21 days. The rheological properties of the CATGs were measured and the incorporation of DNA into the CATGs was assessed. Overall, the CATGs demonstrated increased viscoelastic behavior with the addition of DNA. In addition, two methods of sampling were considered, where it was found that cutting a sample of the dried CATG produced higher allele peak heights in the genotype compared to swabbing. Overall, our CATG biomaterials can be designed for multiple applications in forensic science with tunable properties for various training and research needs.
Collapse
Affiliation(s)
- Amanda Orr
- Environmental and Life Sciences Graduate Program (PhD), Trent University, 1600 West Bank Drive, Peterborough, ON K9J 0G2, Canada.
| | - Paul Wilson
- Biology Department, Trent University, 1600 West Bank Drive, Peterborough, ON K9J 0G2, Canada
| | - Theresa Stotesbury
- Faculty of Science, Forensic Science, Ontario Tech University, 2000 Simcoe Street North, Oshawa, ON L1G 0C5, Canada
| |
Collapse
|
130
|
Wang Y, Wang Q, Luo S, Chen Z, Zheng X, Kankala RK, Chen A, Wang S. 3D bioprinting of conductive hydrogel for enhanced myogenic differentiation. Regen Biomater 2021; 8:rbab035. [PMID: 34408909 PMCID: PMC8363764 DOI: 10.1093/rb/rbab035] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, hydrogels have gained enormous interest in three-dimensional (3D) bioprinting toward developing functional substitutes for tissue remolding. However, it is highly challenging to transmit electrical signals to cells due to the limited electrical conductivity of the bioprinted hydrogels. Herein, we demonstrate the 3D bioprinting-assisted fabrication of a conductive hydrogel scaffold based on poly-3,4-ethylene dioxythiophene (PEDOT) nanoparticles (NPs) deposited in gelatin methacryloyl (GelMA) for enhanced myogenic differentiation of mouse myoblasts (C2C12 cells). Initially, PEDOT NPs are dispersed in the hydrogel uniformly to enhance the conductive property of the hydrogel scaffold. Notably, the incorporated PEDOT NPs showed minimal influence on the printing ability of GelMA. Then, C2C12 cells are successfully encapsulated within GelMA/PEDOT conductive hydrogels using 3D extrusion bioprinting. Furthermore, the proliferation, migration and differentiation efficacies of C2C12 cells in the highly conductive GelMA/PEDOT composite scaffolds are demonstrated using various in vitro investigations of live/dead staining, F-actin staining, desmin and myogenin immunofluorescence staining. Finally, the effects of electrical signals on the stimulation of the scaffolds are investigated toward the myogenic differentiation of C2C12 cells and the formation of myotubes in vitro. Collectively, our findings demonstrate that the fabrication of the conductive hydrogels provides a feasible approach for the encapsulation of cells and the regeneration of the muscle tissue.
Collapse
Affiliation(s)
- Ying Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, P. R. China
| | - Qingshuai Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Shengchang Luo
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Zhoujiang Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Xiang Zheng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Shibin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| |
Collapse
|
131
|
Matsuzaki S. Mechanobiology of the female reproductive system. Reprod Med Biol 2021; 20:371-401. [PMID: 34646066 PMCID: PMC8499606 DOI: 10.1002/rmb2.12404] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Mechanobiology in the field of human female reproduction has been extremely challenging technically and ethically. METHODS The present review provides the current knowledge on mechanobiology of the female reproductive system. This review focuses on the early phases of reproduction from oocyte development to early embryonic development, with an emphasis on current progress. MAIN FINDINGS RESULTS Optimal, well-controlled mechanical cues are required for female reproductive system physiology. Many important questions remain unanswered; whether and how mechanical imbalances among the embryo, decidua, and uterine muscle contractions affect early human embryonic development, whether the biomechanical properties of oocytes/embryos are potential biomarkers for selecting high-quality oocytes/embryos, whether mechanical properties differ between the two major compartments of the ovary (cortex and medulla) in normally ovulating human ovaries, whether durotaxis is involved in several processes in addition to embryonic development. Progress in mechanobiology is dependent on development of technologies that enable precise physical measurements. CONCLUSION More studies are needed to understand the roles of forces and changes in the mechanical properties of female reproductive system physiology. Recent and future technological advancements in mechanobiology research will help us understand the role of mechanical forces in female reproductive system disorders/diseases.
Collapse
Affiliation(s)
- Sachiko Matsuzaki
- CHU Clermont‐FerrandChirurgie GynécologiqueClermont‐FerrandFrance
- Université Clermont AuvergneInstitut Pascal, UMR6602, CNRS/UCA/SIGMAClermont‐FerrandFrance
| |
Collapse
|
132
|
Keskin D, Zu G, Forson AM, Tromp L, Sjollema J, van Rijn P. Nanogels: A novel approach in antimicrobial delivery systems and antimicrobial coatings. Bioact Mater 2021; 6:3634-3657. [PMID: 33898869 PMCID: PMC8047124 DOI: 10.1016/j.bioactmat.2021.03.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
The implementation of nanotechnology to develop efficient antimicrobial systems has a significant impact on the prospects of the biomedical field. Nanogels are soft polymeric particles with an internally cross-linked structure, which behave as hydrogels and can be reversibly hydrated/dehydrated (swollen/shrunken) by the dispersing solvent and external stimuli. Their excellent properties, such as biocompatibility, colloidal stability, high water content, desirable mechanical properties, tunable chemical functionalities, and interior gel-like network for the incorporation of biomolecules, make them fascinating in the field of biological/biomedical applications. In this review, various approaches will be discussed and compared to the newly developed nanogel technology in terms of efficiency and applicability for determining their potential role in combating infections in the biomedical area including implant-associated infections.
Collapse
Affiliation(s)
| | | | | | - Lisa Tromp
- University of Groningen, University Medical Center Groningen, Department of Biomedical Engineering, W. J. Kolff Institute, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Jelmer Sjollema
- University of Groningen, University Medical Center Groningen, Department of Biomedical Engineering, W. J. Kolff Institute, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Patrick van Rijn
- University of Groningen, University Medical Center Groningen, Department of Biomedical Engineering, W. J. Kolff Institute, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| |
Collapse
|
133
|
Zou T, Jiang S, Yi B, Chen Q, Heng BC, Zhang C. Gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor enhances small molecule-induced neurogenic differentiation of stem cells from apical papilla. J Biomed Mater Res A 2021; 110:623-634. [PMID: 34590393 DOI: 10.1002/jbm.a.37315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
The limited neurogenic potential of adult stem cells and their non-specific lineage differentiation pose major challenges in cell-replacement therapy for neurological disorders. In our previous study, we demonstrated that the neurogenic potential of stem cells from apical papilla (SCAPs) was significantly improved upon induction with a small molecule cocktail. This study attempted to investigate whether neuronal differentiation of SCAPs induced by a small molecule cocktail can be further enhanced in a three-dimensional gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor (BDNF-GelMA). The physiological properties and neural differentiation of SCAPs treated with a combination of small molecules and BDNF-GelMA were evaluated by CCK8, Live/Dead assay, quantitative reverse transcription-polymerase chain reaction, western blot and immunocytochemistry. SCAPs embedded in BDNF-GelMA displayed superior morphological characteristics when induced by a small molecule cocktail, similar to neuronal phenotypes as compared to pure GelMA. There was significant upregulation of neural markers including Tuj1 and MAP2 by SCAPs embedded in BDNF-GelMA, as compared to pure GelMA. Hence, GelMA hydrogel loaded with a potent neurotrophic factor (BDNF) provides a conducive scaffold that can further enhance the differentiation of small molecule-treated SCAPs into neuronal-like cells, which may provide a therapeutic platform for the management of neurological disorders.
Collapse
Affiliation(s)
- Ting Zou
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shan Jiang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Baicheng Yi
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qixin Chen
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | | | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
134
|
Paradiso F, Serpelloni S, Francis LW, Taraballi F. Mechanical Studies of the Third Dimension in Cancer: From 2D to 3D Model. Int J Mol Sci 2021; 22:10098. [PMID: 34576261 PMCID: PMC8472581 DOI: 10.3390/ijms221810098] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
From the development of self-aggregating, scaffold-free multicellular spheroids to the inclusion of scaffold systems, 3D models have progressively increased in complexity to better mimic native tissues. The inclusion of a third dimension in cancer models allows researchers to zoom out from a significant but limited cancer cell research approach to a wider investigation of the tumor microenvironment. This model can include multiple cell types and many elements from the extracellular matrix (ECM), which provides mechanical support for the tissue, mediates cell-microenvironment interactions, and plays a key role in cancer cell invasion. Both biochemical and biophysical signals from the extracellular space strongly influence cell fate, the epigenetic landscape, and gene expression. Specifically, a detailed mechanistic understanding of tumor cell-ECM interactions, especially during cancer invasion, is lacking. In this review, we focus on the latest achievements in the study of ECM biomechanics and mechanosensing in cancer on 3D scaffold-based and scaffold-free models, focusing on each platform's level of complexity, up-to-date mechanical tests performed, limitations, and potential for further improvements.
Collapse
Affiliation(s)
- Francesca Paradiso
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Stefano Serpelloni
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| | - Lewis W. Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| |
Collapse
|
135
|
Rana MM, De la Hoz Siegler H. Tuning the Properties of PNIPAm-Based Hydrogel Scaffolds for Cartilage Tissue Engineering. Polymers (Basel) 2021; 13:3154. [PMID: 34578055 PMCID: PMC8467289 DOI: 10.3390/polym13183154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/15/2023] Open
Abstract
Poly(N-isopropylacrylamide) (PNIPAm) is a three-dimensional (3D) crosslinked polymer that can interact with human cells and play an important role in the development of tissue morphogenesis in both in vitro and in vivo conditions. PNIPAm-based scaffolds possess many desirable structural and physical properties required for tissue regeneration, but insufficient mechanical strength, biocompatibility, and biomimicry for tissue development remain obstacles for their application in tissue engineering. The structural integrity and physical properties of the hydrogels depend on the crosslinks formed between polymer chains during synthesis. A variety of design variables including crosslinker content, the combination of natural and synthetic polymers, and solvent type have been explored over the past decade to develop PNIPAm-based scaffolds with optimized properties suitable for tissue engineering applications. These design parameters have been implemented to provide hydrogel scaffolds with dynamic and spatially patterned cues that mimic the biological environment and guide the required cellular functions for cartilage tissue regeneration. The current advances on tuning the properties of PNIPAm-based scaffolds were searched for on Google Scholar, PubMed, and Web of Science. This review provides a comprehensive overview of the scaffolding properties of PNIPAm-based hydrogels and the effects of synthesis-solvent and crosslinking density on tuning these properties. Finally, the challenges and perspectives of considering these two design variables for developing PNIPAm-based scaffolds are outlined.
Collapse
Affiliation(s)
- Md Mohosin Rana
- Biomedical Engineering Graduate Program, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Hector De la Hoz Siegler
- Biomedical Engineering Graduate Program, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
136
|
Sidhu I, Barwe SP, Kiick KL, Kolb EA, Gopalakrishnapillai A. A 3-D hydrogel based system for hematopoietic differentiation and its use in modeling down syndrome associated transient myeloproliferative disorder. Biomater Sci 2021; 9:6266-6281. [PMID: 34369483 PMCID: PMC8570143 DOI: 10.1039/d1bm00442e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Induced pluripotent stem cells (iPSCs) provide an extraordinary tool for disease modeling owing to their potential to differentiate into the desired cell type. The differentiation of iPSCs is typically performed on 2-dimensional monolayers of stromal cell or animal tissue derived extracellular matrices. Recent advancements in disease modeling have utilized iPSCs in 3-dimensional (3D) cultures to study diseases such as muscular dystrophy, cardiomyopathy, and pulmonary fibrosis. However, these approaches are yet to be explored in modeling the hematological malignancies. Transient myeloproliferative disorder (TMD) is a preleukemic stage, which is induced in 10-20% of children with trisomy 21 possessing the pathognomonic mutation in the transcription factor GATA1. In this study, we established a synthetic 3D iPSC culture system for modeling TMD via hematopoietic differentiation of customized iPSCs. A chemically cross-linkable PEG hydrogel decorated with integrin binding peptide was found to be permissive of hematopoietic differentiation of iPSCs. It provided a cost-effective system for the generation of hematopoietic stem and progenitor cells (HSPCs) with higher yield of early HSPCs compared to traditional 2D culture on Matrigel coated dishes. Characterization of the HSPCs produced from the iPSC lines cultured in 3D showed that the erythroid population was reduced whereas the megakaryoid and myeloid populations were significantly increased in GATA1 mutant trisomic line compared to disomic or trisomic lines with wild-type GATA1, consistent with TMD characteristics. In conclusion, we have identified a cost-effective tunable 3D hydrogel system to model TMD.
Collapse
Affiliation(s)
- Ishnoor Sidhu
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
- University of Delaware, Newark, DE 19711, USA
| | - Sonali P Barwe
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
- University of Delaware, Newark, DE 19711, USA
| | | | - E Anders Kolb
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
| | - Anilkumar Gopalakrishnapillai
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
- University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
137
|
Dong YC, Bouché M, Uman S, Burdick JA, Cormode DP. Detecting and Monitoring Hydrogels with Medical Imaging. ACS Biomater Sci Eng 2021; 7:4027-4047. [PMID: 33979137 PMCID: PMC8440385 DOI: 10.1021/acsbiomaterials.0c01547] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hydrogels, water-swollen polymer networks, are being applied to numerous biomedical applications, such as drug delivery and tissue engineering, due to their potential tunable rheologic properties, injectability into tissues, and encapsulation and release of therapeutics. Despite their promise, it is challenging to assess their properties in vivo and crucial information such as hydrogel retention at the site of administration and in situ degradation kinetics are often lacking. To address this, technologies to evaluate and track hydrogels in vivo with various imaging techniques have been developed in recent years, including hydrogels functionalized with contrast generating material that can be imaged with methods such as X-ray computed tomography (CT), magnetic resonance imaging (MRI), optical imaging, and nuclear imaging systems. In this review, we will discuss emerging approaches to label hydrogels for imaging, review the advantages and limitations of these imaging techniques, and highlight examples where such techniques have been implemented in biomedical applications.
Collapse
Affiliation(s)
- Yuxi C Dong
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Mathilde Bouché
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54000 Nancy, France
| | - Selen Uman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David P Cormode
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
138
|
Polonchuk L, Surija L, Lee MH, Sharma P, Liu Chung Ming C, Richter F, Ben-Sefer E, Rad MA, Mahmodi Sheikh Sarmast H, Shamery WA, Tran HA, Vettori L, Haeusermann F, Filipe EC, Rnjak-Kovacina J, Cox T, Tipper J, Kabakova I, Gentile C. Towards engineering heart tissues from bioprinted cardiac spheroids. Biofabrication 2021; 13. [PMID: 34265755 DOI: 10.1088/1758-5090/ac14ca] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
Currentin vivoandin vitromodels fail to accurately recapitulate the human heart microenvironment for biomedical applications. This study explores the use of cardiac spheroids (CSs) to biofabricate advancedin vitromodels of the human heart. CSs were created from human cardiac myocytes, fibroblasts and endothelial cells (ECs), mixed within optimal alginate/gelatin hydrogels and then bioprinted on a microelectrode plate for drug testing. Bioprinted CSs maintained their structure and viability for at least 30 d after printing. Vascular endothelial growth factor (VEGF) promoted EC branching from CSs within hydrogels. Alginate/gelatin-based hydrogels enabled spheroids fusion, which was further facilitated by addition of VEGF. Bioprinted CSs contracted spontaneously and under stimulation, allowing to record contractile and electrical signals on the microelectrode plates for industrial applications. Taken together, our findings indicate that bioprinted CSs can be used to biofabricate human heart tissues for long termin vitrotesting. This has the potential to be used to study biochemical, physiological and pharmacological features of human heart tissue.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- F Hoffmann-La Roche AG Research and Development Division, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel, Basel-Stadt CH-4070, Switzerland
| | - Lydia Surija
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia
| | - Min Ho Lee
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia
| | - Poonam Sharma
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia.,The University of Newcastle Faculty of Health and Medicine, University Drive, Callaghan, NSW 2308, Australia.,University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Clara Liu Chung Ming
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Florian Richter
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia
| | - Eitan Ben-Sefer
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Maryam Alsadat Rad
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Hadi Mahmodi Sheikh Sarmast
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Wafa Al Shamery
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Hien A Tran
- School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Laura Vettori
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Fabian Haeusermann
- F Hoffmann-La Roche AG Research and Development Division, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel, Basel-Stadt CH-4070, Switzerland
| | - Elysse C Filipe
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia.,St Vincent Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | | | - Thomas Cox
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia.,St Vincent Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joanne Tipper
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Irina Kabakova
- University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| | - Carmine Gentile
- The University of Sydney Faculty of Medicine and Health, Kolling Building, Kolling Institute, St Leonards, Sydney, NSW 2065, Australia.,University of Technology Sydney Faculty of Engineering and IT, Building 11, Level 10, Room 115, Ultimo, Sydney, NSW 2007, Australia
| |
Collapse
|
139
|
Ahn W, Lee JH, Kim SR, Lee J, Lee EJ. Designed protein- and peptide-based hydrogels for biomedical sciences. J Mater Chem B 2021; 9:1919-1940. [PMID: 33475659 DOI: 10.1039/d0tb02604b] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Proteins are fundamentally the most important macromolecules for biochemical, mechanical, and structural functions in living organisms. Therefore, they provide us with diverse structural building blocks for constructing various types of biomaterials, including an important class of such materials, hydrogels. Since natural peptides and proteins are biocompatible and biodegradable, they have features advantageous for their use as the building blocks of hydrogels for biomedical applications. They display constitutional and mechanical similarities with the native extracellular matrix (ECM), and can be easily bio-functionalized via genetic and chemical engineering with features such as bio-recognition, specific stimulus-reactivity, and controlled degradation. This review aims to give an overview of hydrogels made up of recombinant proteins or synthetic peptides as the structural elements building the polymer network. A wide variety of hydrogels composed of protein or peptide building blocks with different origins and compositions - including β-hairpin peptides, α-helical coiled coil peptides, elastin-like peptides, silk fibroin, and resilin - have been designed to date. In this review, the structures and characteristics of these natural proteins and peptides, with each of their gelation mechanisms, and the physical, chemical, and mechanical properties as well as biocompatibility of the resulting hydrogels are described. In addition, this review discusses the potential of using protein- or peptide-based hydrogels in the field of biomedical sciences, especially tissue engineering.
Collapse
Affiliation(s)
- Wonkyung Ahn
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Republic of Korea. and Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea.
| | - Jong-Hwan Lee
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Soo Rin Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Jeewon Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea.
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
140
|
King JL, Benhabbour SR. Glioblastoma Multiforme-A Look at the Past and a Glance at the Future. Pharmaceutics 2021; 13:pharmaceutics13071053. [PMID: 34371744 PMCID: PMC8309001 DOI: 10.3390/pharmaceutics13071053] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 01/14/2023] Open
Abstract
Gliomas are the most common type of brain tumor that occur in adults and children. Glioblastoma multiforme (GBM) is the most common, aggressive form of brain cancer in adults and is universally fatal. The current standard-of-care options for GBM include surgical resection, radiotherapy, and concomitant and/or adjuvant chemotherapy. One of the major challenges that impedes success of chemotherapy is the presence of the blood–brain barrier (BBB). Because of the tightly regulated BBB, immune surveillance in the central nervous system (CNS) is poor, contributing to unregulated glioma cell growth. This review gives a comprehensive overview of the latest advances in treatment of GBM with emphasis on the significant advances in immunotherapy and novel therapeutic delivery strategies to enhance treatment for GBM.
Collapse
Affiliation(s)
- Jasmine L. King
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Soumya Rahima Benhabbour
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-843-6142
| |
Collapse
|
141
|
King JL, Benhabbour SR. Glioblastoma Multiforme-A Look at the Past and a Glance at the Future. Pharmaceutics 2021; 13:1053. [PMID: 34371744 PMCID: PMC8309001 DOI: 10.3390/pharmaceutics13071053;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Gliomas are the most common type of brain tumor that occur in adults and children. Glioblastoma multiforme (GBM) is the most common, aggressive form of brain cancer in adults and is universally fatal. The current standard-of-care options for GBM include surgical resection, radiotherapy, and concomitant and/or adjuvant chemotherapy. One of the major challenges that impedes success of chemotherapy is the presence of the blood-brain barrier (BBB). Because of the tightly regulated BBB, immune surveillance in the central nervous system (CNS) is poor, contributing to unregulated glioma cell growth. This review gives a comprehensive overview of the latest advances in treatment of GBM with emphasis on the significant advances in immunotherapy and novel therapeutic delivery strategies to enhance treatment for GBM.
Collapse
Affiliation(s)
- Jasmine L. King
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Soumya Rahima Benhabbour
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-843-6142
| |
Collapse
|
142
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
143
|
Marchini A, Gelain F. Synthetic scaffolds for 3D cell cultures and organoids: applications in regenerative medicine. Crit Rev Biotechnol 2021; 42:468-486. [PMID: 34187261 DOI: 10.1080/07388551.2021.1932716] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Three-dimensional (3D) cell cultures offer an unparalleled platform to recreate spatial arrangements of cells in vitro. 3D cell culture systems have gained increasing interest due to their evident advantages in providing more physiologically relevant information and more predictive data compared to their two-dimensional (2D) counterpart. Design and well-established fabrication of organoids (a particular type of 3D cell culture system) are nowadays considered a pivotal achievement for their ability to replicate in vitro cytoarchitecture and the functionalities of an organ. In this condition, pluripotent stem cells self-organize into 3D structures mimicking the in vivo microenvironments, architectures and multi-lineage differentiation. Scaffolds are key supporting elements in these 3D constructs, and Matrigel is the most commonly used matrix despite its relevant translation limitations including animal-derived sources, non-defined composition, batch-to-batch variability and poorly tailorable properties. Alternatively, 3D synthetic scaffolds, including self-assembling peptides, show promising biocompatibility and biomimetic properties, and can be tailored on specific target tissue/cells. In this review, we discuss the recent advances on 3D cell culture systems and organoids, promising tools for tissue engineering and regenerative medicine applications. For this purpose, we will describe the current state-of-the-art on 3D cell culture systems and organoids based on currently available synthetic-based biomaterials (including tailored self-assembling peptides) either tested in in vivo animal models or developed in vitro with potential application in the field of tissue engineering, with the aim to inspire researchers to take on such promising platforms for clinical applications in the near future.
Collapse
Affiliation(s)
- Amanda Marchini
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Fabrizio Gelain
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
144
|
Han S, Nie K, Li J, Sun Q, Wang X, Li X, Li Q. 3D Electrospun Nanofiber-Based Scaffolds: From Preparations and Properties to Tissue Regeneration Applications. Stem Cells Int 2021; 2021:8790143. [PMID: 34221024 PMCID: PMC8225450 DOI: 10.1155/2021/8790143] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 12/28/2022] Open
Abstract
Electrospun nanofibers have been frequently used for tissue engineering due to their morphological similarities with the extracellular matrix (ECM) and tunable chemical and physical properties for regulating cell behaviors and functions. However, most of the existing electrospun nanofibers have a closely packed two-dimensional (2D) membrane with the intrinsic shortcomings of limited cellular infiltration, restricted nutrition diffusion, and unsatisfied thickness. Three-dimensional (3D) electrospun nanofiber-based scaffolds can provide stem cells with 3D microenvironments and biomimetic fibrous structures. Thus, they have been demonstrated to be good candidates for in vivo repair of different tissues. This review summarizes the recent developments in 3D electrospun nanofiber-based scaffolds (ENF-S) for tissue engineering. Three types of 3D ENF-S fabricated using different approaches classified into electrospun nanofiber 3D scaffolds, electrospun nanofiber/hydrogel composite 3D scaffolds, and electrospun nanofiber/porous matrix composite 3D scaffolds are discussed. New functions for these 3D ENF-S and properties, such as facilitated cell infiltration, 3D fibrous architecture, enhanced mechanical properties, and tunable degradability, meeting the requirements of tissue engineering scaffolds were discovered. The applications of 3D ENF-S in cartilage, bone, tendon, ligament, skeletal muscle, nerve, and cardiac tissue regeneration are then presented with a discussion of current challenges and future directions. Finally, we give summaries and future perspectives of 3D ENF-S in tissue engineering and clinical transformation.
Collapse
Affiliation(s)
- Shanshan Han
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Kexin Nie
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Jingchao Li
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| | - Qingqing Sun
- Center for Functional Sensor and Actuator, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
145
|
Subcutaneously Injectable Hyaluronic Acid Hydrogel for Sustained Release of Donepezil with Reduced Initial Burst Release: Effect of Hybridization of Microstructured Lipid Carriers and Albumin. Pharmaceutics 2021; 13:pharmaceutics13060864. [PMID: 34208289 PMCID: PMC8230846 DOI: 10.3390/pharmaceutics13060864] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
The daily oral administration of acetylcholinesterase (AChE) inhibitors for Alzheimer’s disease features low patient compliance and can lead to low efficacy or high toxicity owing to irregular intake. Herein, we developed a subcutaneously injectable hyaluronic acid hydrogel (MLC/HSA hydrogel) hybridized with microstructured lipid carriers (MLCs) and human serum albumin (HSA) for the sustained release of donepezil (DNP) with reduced initial burst release. The lipid carrier was designed to have a microsized mean diameter (32.6 ± 12.8 µm) to be well-localized in the hydrogel. The hybridization of MLCs and HSA enhanced the structural integrity of the HA hydrogel, as demonstrated by the measurements of storage modulus (G′), loss modulus (G″), and viscosity. In the pharmacokinetic study, subcutaneous administration of MLC/HSA hydrogel in rats prolonged the release of DNP for up to seven days and reduced the initial plasma concentration, where the Cmax value was 0.3-fold lower than that of the control hydrogel without a significant change in the AUClast value. Histological analyses of the hydrogels supported their biocompatibility for subcutaneous injection. These results suggest that a new hybrid MLC/HSA hydrogel could be promising as a subcutaneously injectable controlled drug delivery system for the treatment of Alzheimer’s disease.
Collapse
|
146
|
Trucco D, Sharma A, Manferdini C, Gabusi E, Petretta M, Desando G, Ricotti L, Chakraborty J, Ghosh S, Lisignoli G. Modeling and Fabrication of Silk Fibroin-Gelatin-Based Constructs Using Extrusion-Based Three-Dimensional Bioprinting. ACS Biomater Sci Eng 2021; 7:3306-3320. [PMID: 34101410 DOI: 10.1021/acsbiomaterials.1c00410] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Robotic dispensing-based 3D bioprinting represents one of the most powerful technologies to develop hydrogel-based 3D constructs with enormous potential in the field of regenerative medicine. The optimization of hydrogel printing parameters, proper geometry and internal architecture of the constructs, and good cell viability during the bioprinting process are the essential requirements. In this paper, an analytical model based on the hydrogel rheological properties was developed to predict the extruded filament width in order to maximize the printed structure's fidelity to the design. Viscosity data of two natural hydrogels were imputed to a power-law model to extrapolate the filament width. Further, the model data were validated by monitoring the obtained filament width as the output. Shear stress values occurring during the bioprinting process were also estimated. Human mesenchymal stromal cells (hMSCs) were encapsulated in the silk fibroin-gelatin (G)-based hydrogel, and a 3D bioprinting process was performed to produce cell-laden constructs. Live and dead assay allowed estimating the impact of needle shear stress on cell viability after the bioprinting process. Finally, we tested the potential of hMSCs to undergo chondrogenic differentiation by evaluating the cartilaginous extracellular matrix production through immunohistochemical analyses. Overall, the use of the proposed analytical model enables defining the optimal printing parameters to maximize the fabricated constructs' fidelity to design parameters before the process execution, enabling to achieve more controlled and standardized products than classical trial-and-error approaches in the biofabrication of engineered constructs. Employing modeling systems exploiting the rheological properties of the hydrogels might be a valid tool in the future for guaranteeing high cell viability and for optimizing tissue engineering approaches in regenerative medicine applications.
Collapse
Affiliation(s)
- Diego Trucco
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy.,The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Aarushi Sharma
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology, Hauz Khas, 110016 New Delhi, India
| | - Cristina Manferdini
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Elena Gabusi
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Mauro Petretta
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio RAMSES, Via di Barbiano 1/10, 40136 Bologna, Italy.,RegenHu Ltd., CH-1690 Villaz St. Pierre, Switzerland
| | - Giovanna Desando
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio RAMSES, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Juhi Chakraborty
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology, Hauz Khas, 110016 New Delhi, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology, Hauz Khas, 110016 New Delhi, India
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
147
|
Olăreț E, Bălănucă B, Onaș AM, Ghițman J, Iovu H, Stancu IC, Serafim A. Double-Cross-Linked Networks Based on Methacryloyl Mucin. Polymers (Basel) 2021; 13:1706. [PMID: 34071088 PMCID: PMC8197136 DOI: 10.3390/polym13111706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 01/09/2023] Open
Abstract
Mucin is a glycoprotein with proven potential in the biomaterials field, but its use is still underexploited for such applications. The present work aims to produce a synthesis of methacryloyl mucin single-network (SN) hydrogels and their double-cross-linked-network (DCN) counterparts. Following the synthesis of the mucin methacryloyl derivative, various SN hydrogels are prepared through the photopolymerization of methacrylate bonds, using reaction media with different pH values. The SN hydrogels are converted into DCN systems via supplementary cross-linking in tannic acid aqueous solution. The chemical modification of mucin is described, and the obtained product is characterized; the structural modification of mucin is assessed through FTIR spectroscopy, and the circular dichroism and the isoelectric point of methacryloyl mucin is evaluated. The affinity for aqueous media of both SN and DCN hydrogels is estimated, and the mechanical properties of the systems are assessed, both at macroscale through uniaxial compression and rheology tests and also at microscale through nanoindentation tests.
Collapse
Affiliation(s)
- Elena Olăreț
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
| | - Brîndușa Bălănucă
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
- Department of Organic Chemistry Costin Nenitescu, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania
| | - Andra Mihaela Onaș
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
| | - Jana Ghițman
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
| | - Horia Iovu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, 050094 Bucharest, Romania
| | - Izabela-Cristina Stancu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
- Faculty of Medical Engineering, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania
| | - Andrada Serafim
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Ghe. Polizu Street, 011061 Bucharest, Romania; (E.O.); (B.B.); (A.M.O.); (J.G.); (H.I.); (I.-C.S.)
| |
Collapse
|
148
|
Abstract
The recent advances in bio-integratable electronics are creating new opportunities for investigating and directing biologically significant processes, yet their performance to date is still limited by the inherent physiochemical and signaling mismatches at the heterogeneous interfaces. Hydrogels represent a unique category of materials to bridge the gap between biological and electronic systems because of their structural/functional similarity to biological tissues and design versatility to accommodate cross-system communication. In this review, we discuss the latest progress in the engineering of hydrogel interfaces for bioelectronics development that promotes (1) structural compatibility, where the mechanical and chemical properties of hydrogels can be modulated to achieve coherent, chronically stable biotic-abiotic junctions; and (2) interfacial signal transduction, where the charge and mass transport within the hydrogel mediators can be rationally programmed to condition/amplify the bioderived signals and enhance the electrical/electrochemical coupling. We will further discuss the application of functional hydrogels in complex physiological environments for bioelectronic integration across different scales/biological levels. These ongoing research efforts have the potential to blur the distinction between living systems and artificial electronics, and ultimately decode and regulate biological functioning for both fundamental inquiries and biomedical applications.
Collapse
Affiliation(s)
- Richard Vo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| | | | | |
Collapse
|
149
|
Karami P, Nasrollahzadeh N, Wyss C, O'Sullivan A, Broome M, Procter P, Bourban PE, Moser C, Pioletti DP. An Intrinsically-Adhesive Family of Injectable and Photo-Curable Hydrogels with Functional Physicochemical Performance for Regenerative Medicine. Macromol Rapid Commun 2021; 42:e2000660. [PMID: 33834552 DOI: 10.1002/marc.202000660] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/24/2021] [Indexed: 12/13/2022]
Abstract
Attaching hydrogels to soft internal tissues is crucial for the development of various biomedical devices. Tough sticky hydrogel patches present high adhesion, yet with lack of injectability and the need for treatment of contacting surface. On the contrary, injectable and photo-curable hydrogels are highly attractive owing to their ease of use, flexibility of filling any shape, and their minimally invasive character, compared to their conventional preformed counterparts. Despite recent advances in material developments, a hydrogel that exhibits both proper injectability and sufficient intrinsic adhesion is yet to be demonstrated. Herein, a paradigm shift is proposed toward the design of intrinsically adhesive networks for injectable and photo-curable hydrogels. The bioinspired design strategy not only provides strong adhesive contact, but also results in a wide window of physicochemical properties. The adhesive networks are based on a family of polymeric backbones where chains are modified to be intrinsically adhesive to host tissue and simultaneously form a hydrogel network via a hybrid cross-linking mechanism. With this strategy, adhesion is achieved through a controlled synergy between the interfacial chemistry and bulk mechanical properties. The functionalities of the bioadhesives are demonstrated for various applications, such as tissue adhesives, surgical sealants, or injectable scaffolds.
Collapse
Affiliation(s)
- Peyman Karami
- Laboratory of Biomechanical Orthopedics, EPFL, Lausanne, 1015, Switzerland
| | | | - Céline Wyss
- Laboratory for Processing of Advanced Composites, EPFL, Lausanne, 1015, Switzerland
| | - Aine O'Sullivan
- Laboratory of Biomechanical Orthopedics, EPFL, Lausanne, 1015, Switzerland
| | - Martin Broome
- Department of Maxillofacial Surgery, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Philip Procter
- Applied Materials Science, Department of Engineering Sciences, Uppsala University, Uppsala, 751 03, Sweden
| | | | - Christophe Moser
- Laboratory of Applied Photonics Devices, EPFL, Lausanne, 1015, Switzerland
| | | |
Collapse
|
150
|
Qu L, Dubey N, Ribeiro JS, Bordini EAF, Ferreira JA, Xu J, Castilho RM, Bottino MC. Metformin-loaded nanospheres-laden photocrosslinkable gelatin hydrogel for bone tissue engineering. J Mech Behav Biomed Mater 2021; 116:104293. [PMID: 33588247 PMCID: PMC8275125 DOI: 10.1016/j.jmbbm.2020.104293] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 11/28/2022]
Abstract
The aim of this investigation was to engineer metformin (MF)-loaded mesoporous silica nanospheres (MSNs)-laden gelatin methacryloyl (GelMA) photocrosslinkable hydrogels and test their effects on the mechanical properties, swelling ratio, drug release, cytocompatibility, and osteogenic differentiation of stem cells from human exfoliated deciduous teeth (SHEDs). As-received and carboxylated MSNs (MSNs-COOH) were characterized by scanning and transmission electron microscopies (SEM and TEM), as well as Fourier-transform infrared spectroscopy (FTIR) prior to hydrogel modification. MF-MSNs-COOH were obtained by loading MF into MSNs at a 1:1 mass ratio. Upon MSNs-COOH laden-hydrogels fabrication, the mechanical properties, swelling ratio and MF release were evaluated. SHEDs were seeded on the hydrogels and cytocompatibility was examined. The effects of the MF-MSNs-COOH/GelMA on the osteogenic differentiation of SHEDs were measured by ALP activity, Alizarin Red assay, and Real-time PCR. Statistics were performed using one-way ANOVA (α = 0.05). Morphological (SEM and TEM) analyses of pristine and carboxylated MSNs revealed a mean particle size of 200 nm and 218 nm, respectively. Importantly, an intrinsic nanoporous structure was noticed. Incorporation of MSNs-COOH at 1.5 mg/mL in GelMA led to the highest compressive modulus and swelling ratio. The addition of MSNs-COOH (up to 3 mg/mL) in GelMA did not impact cell viability. The presence of MF in MSNs-COOH/GelMA significantly promoted cell proliferation. Significant upregulation of osteogenic-related genes (except OCN) were seen for modified (MSNs-COOH and MF-MSNs-COOH) hydrogels when compared to GelMA. Altogether, the engineered MF-MSNs-COOH/GelMA shows great promise in craniomaxillofacial applications as an injectable, cell-free and bioactive therapeutics for bone regeneration.
Collapse
Affiliation(s)
- Liu Qu
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Nileshkumar Dubey
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Juliana S Ribeiro
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Restorative Dentistry, School of Dentistry, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Ester A F Bordini
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Jessica A Ferreira
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Jinping Xu
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Rogerio M Castilho
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|