1
|
Tang Y, Levin M, Long OG, Eisenbach CD, Cohen N, Valentine MT. Data-Driven Framework for the Prediction of PEGDA Hydrogel Mechanics. ACS Biomater Sci Eng 2025; 11:259-267. [PMID: 39656140 DOI: 10.1021/acsbiomaterials.4c01762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Poly(ethylene glycol) diacrylate (PEGDA) hydrogels are biocompatible and photo-cross-linkable, with accessible values of elastic modulus ranging from kPa to MPa, leading to their wide use in biomedical and soft material applications. However, PEGDA gels possess complex microstructures, limiting the use of standard polymer theories to describe them. As a result, we lack a foundational understanding of how to relate their composition, processing, and mechanical properties. To address this need, we use a data-driven approach to develop an empirical predictive framework based on high-quality data obtained from uniaxial compression tests and validated using prior data found in the literature. The developed framework accurately predicts the hydrogel shear modulus and the strain-stiffening coefficient using only synthesis parameters, such as the molecular weight and initial concentration of PEGDA, as inputs. These results provide simple and reliable experimental guidelines for precisely controlling both the low-strain and high-strain mechanical responses of PEGDA hydrogels, thereby facilitating their design for various applications.
Collapse
Affiliation(s)
- Yongkui Tang
- Department of Mechanical Engineering, University of California, Santa Barbara, California 93106, United States
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Michal Levin
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Olivia G Long
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
- Materials Department, University of California, Santa Barbara, California 93106, United States
| | - Claus D Eisenbach
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
- Institut für Polymerchemie, University of Stuttgart, Stuttgart D-70569, Germany
| | - Noy Cohen
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Megan T Valentine
- Department of Mechanical Engineering, University of California, Santa Barbara, California 93106, United States
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
2
|
Xu C, Jiang X, Yin W, Lyu L, Tan H, He W, Wang W, Zhou Z, Zhou H, Xu B, Yu Y, Wang Y, Zhou P, Liu Y, Xu J, Gou M, Hu Y. Microgel-encapsulated tetrandrine nanoparticles promote spinal cord repair by sustaining neuroinflammation inhibition. J Mater Chem B 2025; 13:683-694. [PMID: 39620256 DOI: 10.1039/d4tb02080d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Traumatic spinal cord injury (SCI) initiates an intricate secondary injury cascade, characterized by persistent inflammatory responses with neurotoxic microglia and astrocyte activation. Inhibition of neuroinflammation would significantly benefit SCI treatment. Here, tetrandrine with anti-neuroinflammatory activity was delivered into the intrathecal space for SCI treatment. The tetrandrine was encapsulated in MPEG-PDLLA nanoparticles and further loaded into GelMA microgels via a fast 3D printing process based on digital light. After intrathecal injection, the drug-loaded microgels could sustain the release of tetrandrine in the intrathecal space, resulting in efficient repair of the injured spinal cord with recovery of function. Its mechanisms were associated with the modulation of neurotoxic microglia and astrocytes as well as their crosstalk. This work demonstrates a tetrandrine-loaded microgel with potential application in SCI treatment via sustained inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Chongxi Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Xuebing Jiang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wumeng Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Liang Lyu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Huixin Tan
- Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, China
| | - Wenbo He
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Wangyang Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhengyu Zhou
- Clinical Medicine School of Chongqing Medical University, Chongqing Medical University, Chongqing, 401331, China
| | - Hongyu Zhou
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610000, China
| | - Bin Xu
- Neurosurgery Department of West China Tianfu Hospital, Sichuan University, Chengdu, 610000, China
| | - Yang Yu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Peizhi Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Yi Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| | - Maling Gou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yu Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu, Sichuan, 610000, China.
| |
Collapse
|
3
|
Zhang W, Zhao J, Zou X, Yu J, Liao J, Huang F. Multifunctional hydrogels for the healing of oral ulcers. J Biomed Mater Res A 2025; 113:e37776. [PMID: 39210659 DOI: 10.1002/jbm.a.37776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Oral ulcers are one of the most common oral diseases in clinical practice. Its etiology is complex and varied. Due to the dynamic nature of the oral environment, the wound surface is painful due to contact and wear, which seriously affects the quality of life of patients. Oral ulcers are often treated with topical drug therapy. Studies have shown that functional hydrogels play a positive role in promoting wound healing, showing unique advantages in wound dressings. In this paper, the causes and healing characteristics of oral ulcers are discussed in depth, and then the common treatment methods for oral ulcers are summarized and compared. Finally, the potential of functional hydrogels in the treatment of oral ulcers is discussed and projected through a review of the literature in recent years.
Collapse
Affiliation(s)
- Wenjie Zhang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jie Zhao
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinxin Zou
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jingrong Yu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jinlong Liao
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fengjie Huang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
4
|
Zatorski JM, Lee IL, Ortiz-Cárdenas JE, Ellena JF, Pompano RR. Measurement of Covalent Bond Formation in Light-Curing Hydrogels Predicts Physical Stability under Flow. Anal Chem 2024; 96:19880-19888. [PMID: 39625220 DOI: 10.1021/acs.analchem.4c03482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Photo-crosslinking hydrogels are promising for tissue engineering and regenerative medicine, but challenges in reaction monitoring often leave their optimization subject to trial and error. The stability of crosslinked gels under fluid flow, as in the case of a microfluidic device, is particularly challenging to predict, both because of obstacles inherent to solid-state macromolecular analysis that prevent accurate chemical monitoring and because stability is dependent on size of the patterned features. To solve both problems, we obtained 1H NMR spectra of cured hydrogels which were enzymatically degraded. This allowed us to take advantage of the high-resolution that solution NMR provides. This unique approach enabled the measurement of degree of cross-linking (DoC) and prediction of material stability under physiological fluid flow. We showed that NMR spectra of enzyme-digested gels successfully reported on DoC as a function of light exposure and wavelength within two classes of photo-cross-linkable hydrogels: methacryloyl-modified gelatin and a composite of thiol-modified gelatin and norbornene-terminated polyethylene glycol. This approach revealed that a threshold DoC was required for patterned features in each material to become stable and that smaller features required a higher DoC for stability. Finally, we demonstrated that DoC was predictive of the stability of architecturally complex features when photopatterning, underscoring the value of monitoring DoC when using light-reactive gels. We anticipate that the ability to quantify chemical cross-links will accelerate the design of advanced hydrogel materials for structurally demanding applications such as photopatterning and bioprinting.
Collapse
Affiliation(s)
- Jonathan M Zatorski
- Department of Chemistry, University of Virginia, 409 McCormick Road, Charlottesville, Virginia 22904, United States
| | - Isabella L Lee
- Department of Chemistry, University of Virginia, 409 McCormick Road, Charlottesville, Virginia 22904, United States
| | - Jennifer E Ortiz-Cárdenas
- Department of Bioengineering, Stanford University, 443 Via Ortega, Rm 119, Stanford, California 94305, United States
| | - Jeffrey F Ellena
- Department of Chemistry, University of Virginia, 409 McCormick Road, Charlottesville, Virginia 22904, United States
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, 409 McCormick Road, Charlottesville, Virginia 22904, United States
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Sciences, Thornton Hall, 351 McCormick Rd, Charlottesville, Virginia 22904, United States
- Carter Immunology Center and UVA Cancer Center, University of Virginia, 345 Crispell Dr., MR-6, Charlottesville, Virginia 22908, United States
| |
Collapse
|
5
|
Thai NB, Beaman HT, Perlman M, Obeng EE, Du C, Monroe MBB. Chitosan Poly(vinyl alcohol) Methacrylate Hydrogels for Tissue Engineering Scaffolds. ACS APPLIED BIO MATERIALS 2024; 7:7818-7827. [PMID: 38380883 PMCID: PMC11653253 DOI: 10.1021/acsabm.3c01209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/22/2024]
Abstract
A major challenge in tissue engineering scaffolds is controlling scaffold degradation rates during healing while maintaining mechanical properties to support tissue formation. Hydrogels are three-dimensional matrices that are widely applied as tissue scaffolds based on their unique properties that can mimic the extracellular matrix. In this study, we develop a hybrid natural/synthetic hydrogel platform to tune the properties for tissue engineering scaffold applications. We modified chitosan and poly(vinyl alcohol) (PVA) with photo-cross-linkable methacrylate functional groups and then synthesized a library of chitosan PVA methacrylate hydrogels (ChiPVAMA) with two different photoinitiators, Irgacure 2959 (I2959) and lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP). ChiPVAMA hydrogels showed tunability in degradation rates and mechanical properties based on both the polymer content and photoinitiator type. This tunability could enable their application in a range of tissue scaffold applications. In a 2D scratch wound healing assay, all hydrogel samples induced faster wound closure compared to a gauze clinical wound dressing control. NIH/3T3 cells encapsulated in hydrogels showed a high viability (∼92%) over 14 days, demonstrating the capacity of this system as a supportive cell scaffold. In addition, hydrogels containing a higher chitosan content demonstrated a high antibacterial capacity. Overall, ChiPVAMA hydrogels provide a potential tissue engineering scaffold that is tunable, degradable, and suitable for cell growth.
Collapse
Affiliation(s)
- Nghia
Le Ba Thai
- Department
of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute,
and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Henry T. Beaman
- Department
of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute,
and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Megan Perlman
- Department
of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute,
and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Ernest E. Obeng
- Department
of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute,
and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Changling Du
- Department
of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute,
and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Mary Beth B. Monroe
- Department
of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute,
and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
6
|
Hu Y, Zhu T, Cui H, Cui H. Integrating 3D Bioprinting and Organoids to Better Recapitulate the Complexity of Cellular Microenvironments for Tissue Engineering. Adv Healthc Mater 2024:e2403762. [PMID: 39648636 DOI: 10.1002/adhm.202403762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/16/2024] [Indexed: 12/10/2024]
Abstract
Organoids, with their capacity to mimic the structures and functions of human organs, have gained significant attention for simulating human pathophysiology and have been extensively investigated in the recent past. Additionally, 3D bioprinting, as an emerging bio-additive manufacturing technology, offers the potential for constructing heterogeneous cellular microenvironments, thereby promoting advancements in organoid research. In this review, the latest developments in 3D bioprinting technologies aimed at enhancing organoid engineering are introduced. The commonly used bioprinting methods and materials for organoids, with a particular emphasis on the potential advantages of combining 3D bioprinting with organoids are summarized. These advantages include achieving high cell concentrations to form large cellular aggregates, precise deposition of building blocks to create organoids with complex structures and functions, and automation and high throughput to ensure reproducibility and standardization in organoid culture. Furthermore, this review provides an overview of relevant studies from recent years and discusses the current limitations and prospects for future development.
Collapse
Affiliation(s)
- Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tong Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haijun Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
7
|
K Jang L, T Ahlquist J, Ye C, Trujillo J, Triplett M, L Moya M, Robertson C, Hynes W, M Wasson E. Rapid curing dynamics of PEG-thiol-ene resins allow facile 3D bioprinting and in-air cell-laden microgel fabrication. Biomed Mater 2024; 20:015009. [PMID: 39584565 DOI: 10.1088/1748-605x/ad8540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/09/2024] [Indexed: 11/26/2024]
Abstract
Thiol-norbornene photoclick hydrogels are highly efficient in tissue engineering applications due to their fast gelation, cytocompatibility, and tunability. In this work, we utilized the advantageous features of polyethylene glycol (PEG)-thiol-ene resins to enable fabrication of complex and heterogeneous tissue scaffolds using 3D bioprinting and in-air drop encapsulation techniques. We demonstrated that photoclickable PEG-thiol-ene resins could be tuned by varying the ratio of PEG-dithiol to PEG norbornene to generate a wide range of mechanical stiffness (0.5-12 kPa) and swelling ratios. Importantly, all formulations maintained a constant, rapid gelation time (<0.5 s). We used this resin in biological projection microstereolithography (BioPµSL) to print complex structures with geometric fidelity and demonstrated biocompatibility by printing cell-laden microgrids. Moreover, the rapid gelling kinetics of this resin permitted high-throughput fabrication of tunable, cell-laden microgels in air using a biological in-air drop encapsulation apparatus (BioIDEA). We demonstrated that these microgels could support cell viability and be assembled into a gradient structure. This PEG-thiol-ene resin, along with BioPµSL and BioIDEA technology, will allow rapid fabrication of complex and heterogeneous tissues that mimic native tissues with cellular and mechanical gradients. The engineered tissue scaffolds with a controlled microscale porosity could be utilized in applications including gradient tissue engineering, biosensing, andin vitrotissue models.
Collapse
Affiliation(s)
- Lindy K Jang
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Jesse T Ahlquist
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Congwang Ye
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Juliana Trujillo
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
| | - Michael Triplett
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| | - William Hynes
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Elisa M Wasson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| |
Collapse
|
8
|
Lai Y, Xiao X, Huang Z, Duan H, Yang L, Yang Y, Li C, Feng L. Photocrosslinkable Biomaterials for 3D Bioprinting: Mechanisms, Recent Advances, and Future Prospects. Int J Mol Sci 2024; 25:12567. [PMID: 39684279 DOI: 10.3390/ijms252312567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Constructing scaffolds with the desired structures and functions is one of the main goals of tissue engineering. Three-dimensional (3D) bioprinting is a promising technology that enables the personalized fabrication of devices with regulated biological and mechanical characteristics similar to natural tissues/organs. To date, 3D bioprinting has been widely explored for biomedical applications like tissue engineering, drug delivery, drug screening, and in vitro disease model construction. Among different bioinks, photocrosslinkable bioinks have emerged as a powerful choice for the advanced fabrication of 3D devices, with fast crosslinking speed, high resolution, and great print fidelity. The photocrosslinkable biomaterials used for light-based 3D printing play a pivotal role in the fabrication of functional constructs. Herein, this review outlines the general 3D bioprinting approaches related to photocrosslinkable biomaterials, including extrusion-based printing, inkjet printing, stereolithography printing, and laser-assisted printing. Further, the mechanisms, advantages, and limitations of photopolymerization and photoinitiators are discussed. Next, recent advances in natural and synthetic photocrosslinkable biomaterials used for 3D bioprinting are highlighted. Finally, the challenges and future perspectives of photocrosslinkable bioinks and bioprinting approaches are envisaged.
Collapse
Affiliation(s)
- Yushang Lai
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiong Xiao
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziwei Huang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongying Duan
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liping Yang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuchu Yang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenxi Li
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Feng
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Zhang IW, Choi LS, Friend NE, McCoy AJ, Midekssa FS, Alsberg E, Lesher-Pérez SC, Stegemann JP, Baker BM, Putnam AJ. Clickable PEG-norbornene microgels support suspension bioprinting and microvascular assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623424. [PMID: 39605682 PMCID: PMC11601470 DOI: 10.1101/2024.11.15.623424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The development of perfusable and multiscale vascular networks remains one of the largest challenges in tissue engineering. As such, there is a need for the creation of customizable and facile methods to produce robustly vascularized constructs. In this study, secondarily crosslinkable (clickable) poly(ethylene glycol)-norbornene (PEGNB) microbeads were produced and evaluated for their ability to sequentially support suspension bioprinting and microvascular self-assembly towards the aim of engineering hierarchical vasculature. The clickable PEGNB microbead slurry exhibited mechanical behavior suitable for suspension bioprinting of sacrificial bioinks, could be UV crosslinked into a granular construct post-print, and withstood evacuation of the bioink and subsequent perfusion of the patterned void space. Endothelial and stromal cells co-embedded within jammed RGD-modified PEGNB microbead slurries assembled into capillary-scale vasculature after secondary crosslinking of the beads into granular constructs, with endothelial tubules forming within the interstitial space between microbeads and supported by the perivascular association of the stromal cells. Microvascular self-assembly was not impacted by printing sacrificial bioinks into the cell-laden microbead support bath before UV crosslinking. Collectively, these results demonstrate that clickable PEGNB microbeads are a versatile substrate for both suspension printing and microvascular culture and may be the foundation for a promising methodology to engineer hierarchical vasculature.
Collapse
|
10
|
Joshi V, Singh N, Datta P. Facile fabrication of degradable, serrated polyethylene diacrylate microneedles using stereolithography. Pharm Dev Technol 2024; 29:976-986. [PMID: 39364615 DOI: 10.1080/10837450.2024.2413146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024]
Abstract
Microneedles have the potential for minimally invasive drug delivery. However, they are constrained by absence of rapid, scalable fabrication methods to produce intricate arrays and serrations for enhanced adhesion. 3D printing techniques like stereolithography (SLA) are fast, scalable modalities but SLAs require non-degradable and stiff resins. This work attempts to overcome this limitation by utilizing a poly (ethylene glycol diacrylate) (PEGDA, F3) resin and demonstrating its compatibility with a commercial SLA printer. FESEM images showed high printing efficiency of customized bioinks (F3) similar to commercial resins using SLA 3D printer. Mechanical endurance tests of whole MNA showed that MNs array printed from F3 resin (485 ± 5.73 N) required considerably less force than commercial F1 resin (880 ± 32.4 N). Penetration performance of F1 and F3 was found to be 10.8 ± 2.06 N and 0.705 ± 0.03 N. In-vitro degradation study in PBS showed that MNs fabricated from F3 resin exhibited degradation after 7 days, which was not observed with the commercial F1 resin provided by the manufacturer. The histology of porcine skin exhibited formation of triangular pores with pore length of 548 μm and efficient penetration into the deeper dermal layer. In conclusion, PEGDA can be used as for fabricating degradable, serrated solid MNs over commercial resin.
Collapse
Affiliation(s)
- Vedant Joshi
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, WB, India
| | - Nidhi Singh
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, WB, India
| | - Pallab Datta
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, WB, India
| |
Collapse
|
11
|
Singh A, Cho YK, Cohen DJ. Rapid Whole-Plate Cell and Tissue Micropatterning Using a Budget 3D Resin Printer. ACS OMEGA 2024; 9:43808-43816. [PMID: 39494000 PMCID: PMC11525498 DOI: 10.1021/acsomega.4c06539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024]
Abstract
The ability to precisely pattern cells and proteins is crucial in various scientific disciplines, including cell biology, bioengineering, and materials chemistry. Current techniques, such as microcontact stamping, 3D bioprinting, and direct photopatterning, have limitations in terms of cost, versatility, and throughput. In this Article, we present an accessible approach that combines the throughput of photomask systems with the versatility of programmable light patterning using a low-cost consumer LCD resin printer. The method involves utilizing a bioinert hydrogel, poly(ethylene glycol) diacrylate (PEGDA), and a 405 nm sensitive photoinitiator (LAP) that are selectively cross-linked to form a hydrogel upon light exposure, creating specific regions that are protein and cell-repellent. Our result highlights that a low-cost LCD resin printer can project virtual photomasks onto the hydrogel, allowing for reasonable resolution and large-area printing at a fraction of the cost of traditional systems. The study demonstrates the calibration of exposure times for optimal resolution and accuracy and shape corrections to overcome the inherent challenges of wide-field resin printing. The potential of this approach is validated through widely studied 2D and 3D stem cell applications, showcasing its biocompatibility and ability to replicate complex tissue engineering patterns. We also validate the method with a cell-adhesive polymer (gelatin methacrylate; GelMA). The combination of low cost, high throughput, and accessibility makes this method broadly applicable across fields for enabling rapid and precise fabrication of cells and tissues in standard laboratory culture vessels.
Collapse
Affiliation(s)
- Anamika Singh
- Department
of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Youn Kyoung Cho
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Daniel J. Cohen
- Department
of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
12
|
Dúzs B, Skarsetz O, Fusi G, Lupfer C, Walther A. Mechano-adaptive meta-gels through synergistic chemical and physical information-processing. Nat Commun 2024; 15:8957. [PMID: 39419997 PMCID: PMC11487081 DOI: 10.1038/s41467-024-53368-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Global functional adaptation after local mechanical stimulation, as in mechanobiology and the mimosa plant, is fascinating and ubiquitous in nature. This is achieved by locally sensing mechanical deformation with precise thresholds, processing this information via biochemical circuits, followed by downstream actuation. The integration of such embodied intelligence allowing for mechano-to-chemo-to-function information-processing remains elusive in man-made systems. By merging the fields of chemical circuits and metamaterials, we introduce adaptive metamaterial hydrogels (meta-gels) that can accurately sense mechanical stimuli (local touch and global strain), transmit this information over long distances via reaction-diffusion signaling, and induce downstream mechanical strengthening by growing nanofibril networks, or soft robotic actuation through competitive swelling. All elements of the sensor-processor-actuator system are embedded in the device, functioning autonomously without external feeding reservoirs. Our concept enables designing advanced life-like materials systems that synergistically combine two worlds - chemical circuits for chemical information-processing and metamaterial unit cells for physical information-processing.
Collapse
Affiliation(s)
- Brigitta Dúzs
- Life-Like Materials and Systems, University of Mainz, Duesbergweg 10-14, Mainz, 55128, Germany.
| | - Oliver Skarsetz
- Life-Like Materials and Systems, University of Mainz, Duesbergweg 10-14, Mainz, 55128, Germany
| | - Giorgio Fusi
- Life-Like Materials and Systems, University of Mainz, Duesbergweg 10-14, Mainz, 55128, Germany
| | - Claudius Lupfer
- Life-Like Materials and Systems, University of Mainz, Duesbergweg 10-14, Mainz, 55128, Germany
| | - Andreas Walther
- Life-Like Materials and Systems, University of Mainz, Duesbergweg 10-14, Mainz, 55128, Germany.
| |
Collapse
|
13
|
Korzhikov-Vlakh V, Mikhailova A, Sinitsyna E, Korzhikova-Vlakh E, Tennikova T. Gradient Functionalization of Poly(lactic acid)-Based Materials with Polylysine for Spatially Controlled Cell Adhesion. Polymers (Basel) 2024; 16:2888. [PMID: 39458716 PMCID: PMC11511340 DOI: 10.3390/polym16202888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The development of biomaterials with gradient surface modification capable of spatially controlled cell adhesion and migration is of great importance for tissue engineering and regeneration. In this study, we proposed a method for the covalent modification of PLA-based materials with a cationic polypeptide (polylysine, PLys) via a thiol-ene click reaction carried out under a light gradient. With this aim, PLA-based films were fabricated and modified with 2-aminoethyl methacrylate (AEMA) as a double bond source. The latter was introduced by reacting pre-formed and activated surface carboxyl groups with the amino group of AEMA. The success of the modification was confirmed by 1H NMR, Raman and X-ray photoelectron spectroscopy data. A further photoinduced thiol-ene click reaction in the presence of a photosensitive initiator as a radical source was further optimized using cysteine. For grafting of PLys via the thiol-ene click reaction, PLys with a terminal thiol group was synthesized by ring-opening polymerization using Cys(Acm) as an amine initiator. Deprotection of the polypeptide resulted in the formation of free thiol groups of Cys-PLys. Successful gradient grafting of Cys-PLys was evidenced by covalent staining with the fluorescent dye Cy3-NHS. In addition, PLys gradient-dependent adhesion and migration of HEK 293 cells on PLys-PLA-based surfaces was confirmed.
Collapse
Affiliation(s)
- Viktor Korzhikov-Vlakh
- Institute of Chemistry, St. Petersburg, St. Petersburg State University, 198504 St. Petersburg, Russia; (A.M.); (E.S.); (T.T.)
- St. Petersburg State University Hospital, 199034 St. Petersburg, Russia
| | - Aleksandra Mikhailova
- Institute of Chemistry, St. Petersburg, St. Petersburg State University, 198504 St. Petersburg, Russia; (A.M.); (E.S.); (T.T.)
- Federal Research Center N. I. Vavilov All-Russian Institute of Plant Genetic Resources (VIR), 190000 St. Petersburg, Russia
| | - Ekaterina Sinitsyna
- Institute of Chemistry, St. Petersburg, St. Petersburg State University, 198504 St. Petersburg, Russia; (A.M.); (E.S.); (T.T.)
| | - Evgenia Korzhikova-Vlakh
- Institute of Chemistry, St. Petersburg, St. Petersburg State University, 198504 St. Petersburg, Russia; (A.M.); (E.S.); (T.T.)
| | - Tatiana Tennikova
- Institute of Chemistry, St. Petersburg, St. Petersburg State University, 198504 St. Petersburg, Russia; (A.M.); (E.S.); (T.T.)
| |
Collapse
|
14
|
Boretti G, Amirfallah A, Edmunds KJ, Hamzehpour H, Sigurjónsson ÓE. Advancing Cartilage Tissue Engineering: A Review of 3D Bioprinting Approaches and Bioink Properties. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39381849 DOI: 10.1089/ten.teb.2024.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Articular cartilage is crucial in human physiology, and its degeneration poses a significant public health challenge. While recent advancements in 3D bioprinting and tissue engineering show promise for cartilage regeneration, there remains a gap between research findings and clinical application. This review critically examines the mechanical and biological properties of hyaline cartilage, along with current 3D manufacturing methods and analysis techniques. Moreover, we provide a quantitative synthesis of bioink properties used in cartilage tissue engineering. After screening 181 initial works, 33 studies using extrusion bioprinting were analyzed and synthesized, presenting results that indicate the main materials, cells, and methods utilized for mechanical and biological evaluation. Altogether, this review motivates the standardization of mechanical analyses and biomaterial assessments of 3D bioprinted constructs to clarify their chondrogenic potential.
Collapse
Affiliation(s)
- Gabriele Boretti
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
| | - Arsalan Amirfallah
- The Blood Bank, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| | - Kyle J Edmunds
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
| | - Helena Hamzehpour
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Ólafur E Sigurjónsson
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
- The Blood Bank, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| |
Collapse
|
15
|
Perera KDC, Boiani SM, Vasta AK, Messenger KJ, Delva S, Menon JU. Development and characterization of a novel poly( N-isopropylacrylamide)-based thermoresponsive photoink and its applications in DLP bioprinting. J Mater Chem B 2024; 12:9767-9779. [PMID: 39230440 PMCID: PMC11373533 DOI: 10.1039/d4tb00682h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
The field of 3-dimensional (3D) bioprinting has significantly expanded capabilities in producing precision-engineered hydrogel constructs, and recent years have seen the development of various stimuli-responsive bio- and photoinks. There is, however, a distinct lack of digital light processing (DLP)-compatible photoinks with thermoresponsivity. To remedy this, this work focuses on formulating and optimizing a versatile ink for DLP printing of thermoresponsive hydrogels, with numerous potential applications in tissue engineering, drug delivery, and adjacent biomedical fields. Photoink optimization was carried out using a multifactorial study design. The optimized photoink yielded crosslinked hydrogels with strong variations in hydrophobicity (contact angles of 44.4° LCST), indicating marked thermoresponsivity. Mechanical- and rheological characterization of the printed hydrogels showed significant changes above the LCST: storage- and loss moduli both increased and loss tangent and compressive modulus decreased above this temperature (P ≤ 0.01). The highly cytocompatible hydrogel microwell arrays yielded both single- and multilayer spheroids with human dermal fibroblasts (HDFs) and HeLa cells successfully. Evaluation of the release of encapsulated model macro- (bovine serum albumin, BSA) and small molecule (rhodamine B) drugs in a buffer solution showed an interestingly inverted thermoresponsive release profile with >80% release at room temperature and about 50-60% release above the gels' LCST. All told, the optimized ink holds great promise for multiple biomedical applications including precise and high-resolution fabrication of complex tissue structures, development of smart drug delivery systems and 3D cell culture.
Collapse
Affiliation(s)
- Kalindu D C Perera
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Sophia M Boiani
- Department of Chemical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Alexandra K Vasta
- Department of Chemical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Katherine J Messenger
- Department of Biomedical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Sabrina Delva
- Department of Biomedical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Chemical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
16
|
Baghdasaryan O, Lee-Kin J, Tan C. Architectural engineering of Cyborg Bacteria with intracellular hydrogel. Mater Today Bio 2024; 28:101226. [PMID: 39328785 PMCID: PMC11426140 DOI: 10.1016/j.mtbio.2024.101226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/09/2024] [Accepted: 09/01/2024] [Indexed: 09/28/2024] Open
Abstract
Synthetic biology primarily uses genetic engineering to control living cells. In contrast, recent work has ushered in the architectural engineering of living cells through intracellular materials. Specifically, Cyborg Bacteria are created by incorporating synthetic PEG-based hydrogel inside cells. Cyborg Bacteria do not replicate but maintain essential cellular functions, including metabolism and protein synthesis. Thus far, Cyborg Bacteria have been engineered using one primary composition of intracellular hydrogel components. Here, we demonstrate the versatility of controlling the physical and biochemical aspects of Cyborg Bacteria using different structures of hydrogels. The intracellular cell-gel architecture is modulated using a different photoinitiator, PEG-diacrylate (PEG-DA) of different molecular weights, 4arm PEG-DA, and dsDNA-PEG. We show that the molecular weight of the PEG-DA affects the generation and metabolism of Cyborg Bacteria. In addition, we show that the hybrid dsDNA-PEG intracellular hydrogel controls protein expression levels of the Cyborg Bacteria through post-transcriptional regulation and polymerase sequestration. Our work creates a new frontier of modulating intracellular gel components to control Cyborg Bacteria function and architecture.
Collapse
Affiliation(s)
| | - Jared Lee-Kin
- Biomedical Engineering, University of California Davis, United States
| | - Cheemeng Tan
- Biomedical Engineering, University of California Davis, United States
| |
Collapse
|
17
|
Deo KA, Murali A, Tronolone JJ, Mandrona C, Lee HP, Rajput S, Hargett SE, Selahi A, Sun Y, Alge DL, Jain A, Gaharwar AK. Granular Biphasic Colloidal Hydrogels for 3D Bioprinting. Adv Healthc Mater 2024; 13:e2303810. [PMID: 38749006 DOI: 10.1002/adhm.202303810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/01/2024] [Indexed: 05/30/2024]
Abstract
Granular hydrogels composed of hydrogel microparticles are promising candidates for 3D bioprinting due to their ability to protect encapsulated cells. However, to achieve high print fidelity, hydrogel microparticles need to jam to exhibit shear-thinning characteristics, which is crucial for 3D printing. Unfortunately, this overpacking can significantly impact cell viability, thereby negating the primary advantage of using hydrogel microparticles to shield cells from shear forces. To overcome this challenge, a novel solution: a biphasic, granular colloidal bioink designed to optimize cell viability and printing fidelity is introduced. The biphasic ink consists of cell-laden polyethylene glycol (PEG) hydrogel microparticles embedded in a continuous gelatin methacryloyl (GelMA)-nanosilicate colloidal network. Here, it is demonstrated that this biphasic bioink offers outstanding rheological properties, print fidelity, and structural stability. Furthermore, its utility for engineering complex tissues with multiple cell types and heterogeneous microenvironments is demonstrated, by incorporating β-islet cells into the PEG microparticles and endothelial cells in the GelMA-nanosilicate colloidal network. Using this approach, it is possible to induce cell patterning, enhance vascularization, and direct cellular function. The proposed biphasic bioink holds significant potential for numerous emerging biomedical applications, including tissue engineering and disease modeling.
Collapse
Affiliation(s)
- Kaivalya A Deo
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Aparna Murali
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - James J Tronolone
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Cole Mandrona
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Hung Pang Lee
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Satyam Rajput
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Sarah E Hargett
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Amirali Selahi
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Yuxiang Sun
- Nutrition, College of Agriculture, Texas A&M University, College Station, TX, 77843, USA
| | - Daniel L Alge
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Abhishek Jain
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Medical Physiology, School of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
- Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Akhilesh K Gaharwar
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Interdisciplinary Graduate Program in Genetics & Genomics, Texas A&M University, College Station, TX, 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
18
|
Nelson BR, Kirkpatrick BE, Miksch CE, Davidson MD, Skillin NP, Hach GK, Khang A, Hummel SN, Fairbanks BD, Burdick JA, Bowman CN, Anseth KS. Photoinduced Dithiolane Crosslinking for Multiresponsive Dynamic Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2211209. [PMID: 36715698 PMCID: PMC10387131 DOI: 10.1002/adma.202211209] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/11/2023] [Indexed: 06/18/2023]
Abstract
While many hydrogels are elastic networks crosslinked by covalent bonds, viscoelastic hydrogels with adaptable crosslinks are increasingly being developed to better recapitulate time and position-dependent processes found in many tissues. In this work, 1,2-dithiolanes are presented as dynamic covalent photocrosslinkers of hydrogels, resulting in disulfide bonds throughout the hydrogel that respond to multiple stimuli. Using lipoic acid as a model dithiolane, disulfide crosslinks are formed under physiological conditions, enabling cell encapsulation via an initiator-free light-induced dithiolane ring-opening photopolymerization. The resulting hydrogels allow for multiple photoinduced dynamic responses including stress relaxation, stiffening, softening, and network functionalization using a single chemistry, which can be supplemented by permanent reaction with alkenes to further control network properties and connectivity using irreversible thioether crosslinks. Moreover, complementary photochemical approaches are used to achieve rapid and complete sample degradation via radical scission and post-gelation network stiffening when irradiated in the presence of reactive gel precursor. The results herein demonstrate the versatility of this material chemistry to study and direct 2D and 3D cell-material interactions. This work highlights dithiolane-based hydrogel photocrosslinking as a robust method for generating adaptable hydrogels with a range of biologically relevant mechanical and chemical properties that are varied on demand.
Collapse
Affiliation(s)
- Benjamin R Nelson
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Bruce E Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Medical Scientist Training Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Connor E Miksch
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Matthew D Davidson
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Nathaniel P Skillin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Medical Scientist Training Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Grace K Hach
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Alex Khang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Sydney N Hummel
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Benjamin D Fairbanks
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Jason A Burdick
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Christopher N Bowman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
19
|
Lambrecht S, Gazizova A, Kara S, Meyer J, Jopp S. Antimicrobial properties and biocompatibility of semi-synthetic carbohydrate-based ionic hydrogels. RSC Adv 2024; 14:30719-30731. [PMID: 39328876 PMCID: PMC11425042 DOI: 10.1039/d4ra05695g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Hydrogels have gained significant interest in the last decades, especially in the medical sector, due to their versatile properties. While hydrogels from naturally occurring polysaccharides (e.g. cellulose) are well-known, those produced from polymerizable carbohydrate-based monomers remain underexplored. However, these semi-synthetic hydrogels offer the great advantage of having adjustable properties for customization depending on their application. The objective of this study was to characterize semi-synthetic carbohydrate-based ionic hydrogels produced from GVIM-I (glucosyl vinyl imidazolium iodide). The antimicrobial activity was evaluated using the disk diffusion method, which demonstrated that all samples exhibit inhibitory effects on the growth of Candida auris. In vitro biocompatibility was determined by cell viability studies with L929 mouse fibroblasts, and a correlation was observed between eluate concentration and cell viability. In particular, the type of initiator system employed for polymerization was found to affect cell viability. The direct contact assessments showed that specific pre-treatments of the hydrogels resulted in higher cell viability than non-treated hydrogels. The results also revealed the impact of crosslinker concentration and type and identified poly(ethylene glycol)diacrylate (PEGDA) 575 as a promising crosslinker for future medical applications. LC-MS analysis of the wash medium identified unreacted GVIM-I as the leached material, which is presumed to be the cause of the observed cytotoxicity. Overall, the study provides valuable insights into the characteristics of GVIM-I based hydrogels and sheds light on the factors that influence their cytotoxicity and potential for medical application.
Collapse
Affiliation(s)
- Sina Lambrecht
- Department Life, Light & Matter, University of Rostock Albert-Einstein-Str. 25 18059 Rostock Germany
| | - Alina Gazizova
- Institute of Chemistry, University of Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
| | - Selin Kara
- Institute of Technical Chemistry, Leibniz University Hannover Callinstraße 5 30167 Hannover Germany
- Biocatalysis and Bioprocessing Group, Department of Biological and Chemical Engineering, Aarhus University Gustav Wieds Vej 10 8000 Aarhus Denmark
| | - Johanna Meyer
- Institute of Technical Chemistry, Leibniz University Hannover Callinstraße 5 30167 Hannover Germany
| | - Stefan Jopp
- Department Life, Light & Matter, University of Rostock Albert-Einstein-Str. 25 18059 Rostock Germany
| |
Collapse
|
20
|
Meinen S, Brinkmann S, Viebrock K, Elbardisy B, Menzel H, Krull R, Dietzel A. 2PP-Hydrogel Covered Electrodes to Compensate for Media Effects in the Determination of Biomass in a Capillary Wave Micro Bioreactor. BIOSENSORS 2024; 14:438. [PMID: 39329813 PMCID: PMC11429511 DOI: 10.3390/bios14090438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Microbioreactors increase information output in biopharmaceutical screening applications because they can be operated in parallel without consuming large quantities of the pharmaceutical formulations being tested. A capillary wave microbioreactor (cwMBR) has recently been reported, allowing cost-efficient parallelization in an array that can be activated for mixing as a whole. Although impedance spectroscopy can directly distinguish between dead and viable cells, the monitoring of cells in suspension within bioreactors is challenging because the signal is influenced by the potentially varying properties of the culture medium. In order to address this challenge, an impedance sensor consisting of two sets of microelectrodes in a cwMBR is presented. Only one set of electrodes was covered by a two-photon cross-linked hydrogel to become insensitive to the influence of cells while remaining sensitive to the culture medium. With this impedance sensor, the biomass of Saccharomyces cerevisiae could be measured in a range from 1 to 20 g L-1. In addition, the sensor can compensate for a change in the conductivity of the suspension of 5 to 15 mS cm-1. Moreover, the two-photon cross-linking of hydroxyethyl starch methacrylate hydrogel, which has been studied in detail, recommends itself for even much broader sensing applications in miniaturized bioreactors and biosensors.
Collapse
Affiliation(s)
- Sven Meinen
- Institute of Microtechnology, Technische Universität Braunschweig, Alte Salzdahlumer Str. 203, 38124 Braunschweig, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
| | - Steffen Brinkmann
- Institute of Microtechnology, Technische Universität Braunschweig, Alte Salzdahlumer Str. 203, 38124 Braunschweig, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
| | - Kevin Viebrock
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Bassant Elbardisy
- Institute of Technical Chemistry, Technische Universität Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Henning Menzel
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
- Institute of Technical Chemistry, Technische Universität Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Rainer Krull
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
- Institute of Biochemical Engineering, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Andreas Dietzel
- Institute of Microtechnology, Technische Universität Braunschweig, Alte Salzdahlumer Str. 203, 38124 Braunschweig, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
| |
Collapse
|
21
|
Antczak LAM, Moore KN, Hendrick TE, Heise RL. Binary fabrication of decellularized lung extracellular matrix hybridgels for in vitro chronic obstructive pulmonary disease modeling. Acta Biomater 2024; 185:190-202. [PMID: 39059731 PMCID: PMC11474825 DOI: 10.1016/j.actbio.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
Limited treatments and a lack of appropriate animal models have spurred the study of scaffolds to mimic lung disease in vitro. Decellularized human lung and its application in extracellular matrix (ECM) hydrogels has advanced the development of these lung ECM models. Controlling the biochemical and mechanical properties of decellularized ECM hydrogels continues to be of interest due to inherent discrepancies of hydrogels when compared to their source tissue. To optimize the physiologic relevance of ECM hydrogel lung models without sacrificing the native composition we engineered a binary fabrication system to produce a Hybridgel composed of an ECM hydrogel reinforced with an ECM cryogel. Further, we compared the effect of ECM-altering disease on the properties of the gels using elastin poor Chronic Obstructive Pulmonary Disease (COPD) vs non-diseased (ND) human lung source tissue. Nanoindentation confirmed the significant loss of elasticity in hydrogels compared to that of ND human lung and further demonstrated the recovery of elastic moduli in ECM cryogels and Hybridgels. These findings were supported by similar observations in diseased tissue and gels. Successful cell encapsulation, distribution, cytotoxicity, and infiltration were observed and characterized via confocal microscopy. Cells were uniformly distributed throughout the Hybridgel and capable of survival for 7 days. Cell-laden ECM hybridgels were found to have elasticity similar to that of ND human lung. Compositional investigation into diseased and ND gels indicated the conservation of disease-specific elastin to collagen ratios. In brief, we have engineered a composited ECM hybridgel for the 3D study of cell-matrix interactions of varying lung disease states that optimizes the application of decellularized lung ECM materials to more closely mimic the human lung while conserving the compositional bioactivity of the native ECM. STATEMENT OF SIGNIFICANCE: The lack of an appropriate disease model for the study of chronic lung diseases continues to severely inhibit the advancement of treatments and preventions of these otherwise fatal illnesses due to the inability to recapture the biocomplexity of pathologic cell-ECM interactions. Engineering biomaterials that utilize decellularized lungs offers an opportunity to deconstruct, understand, and rebuild models that highlight and investigate how disease specific characteristics of the extracellular environment are involved in driving disease progression. We have advanced this space by designing a binary fabrication system for a ECM Hybridgel that retains properties from its source material required to observe native matrix interactions. This design simulates a 3D lung environment that is both mechanically elastic and compositionally relevant when derived from non-diseased tissue and pathologically diminished both mechanically and compositionally when derived from COPD tissue. Here we describe the ECM hybridgel as a model for the study of cell-ECM interactions involved in COPD.
Collapse
Affiliation(s)
- Leigh-Ann M Antczak
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Karah N Moore
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Taylor E Hendrick
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, USA.
| |
Collapse
|
22
|
Xiang Y, Gao Y, Cheng Q, Lei Z, Zhang X, Yang Y, Zhang J. Recombinant collagen coating 3D printed PEGDA hydrogel tube loading with differentiable BMSCs to repair bile duct injury. Colloids Surf B Biointerfaces 2024; 241:114064. [PMID: 38954937 DOI: 10.1016/j.colsurfb.2024.114064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Bile duct injury presents a significant clinical challenge following hepatobiliary surgery, necessitating advancements in the repair of damaged bile ducts is a persistent issue in biliary surgery. 3D printed tubular scaffolds have emerged as a promising approach for the repair of ductal tissues, yet the development of scaffolds that balance exceptional mechanical properties with biocompatibility remains an ongoing challenge. This study introduces a novel, bio-fabricated bilayer bile duct scaffold using a 3D printing technique. The scaffold comprises an inner layer of polyethylene glycol diacrylate (PEGDA) to provide high mechanical strength, and an outer layer of biocompatible, methacryloylated recombinant collagen type III (rColMA) loaded with basic fibroblast growth factor (bFGF)-encapsulated liposomes (bFGF@Lip). This design enables the controlled release of bFGF, creating an optimal environment for the growth and differentiation of bone marrow mesenchymal stem cells (BMSCs) into cholangiocyte-like cells. These cells are instrumental in the regeneration of bile duct tissues, evidenced by the pronounced expression of cholangiocyte differentiation markers CK19 and CFTR. The PEGDA//rColMA/bFGF@Lip bilayer bile duct scaffold can well simulate the bile duct structure, and the outer rColMA/bFGF@Lip hydrogel can well promote the growth and differentiation of BMSCs into bile duct epithelial cells. In vivo experiments showed that the scaffold did not cause cholestasis in the body. This new in vitro pre-differentiated active 3D printed scaffold provides new ideas for the study of bile duct tissue replacement.
Collapse
Affiliation(s)
- Yang Xiang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Qiuhua Cheng
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Zhongwen Lei
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Xiaoyu Zhang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Yijun Yang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China.
| | - Jianquan Zhang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China.
| |
Collapse
|
23
|
Pourmostafa A, Bhusal A, Haridas Menon N, Li Z, Basuray S, Miri AK. Integrating conductive electrodes into hydrogel-based microfluidic chips for real-time monitoring of cell response. Front Bioeng Biotechnol 2024; 12:1421592. [PMID: 39257447 PMCID: PMC11384590 DOI: 10.3389/fbioe.2024.1421592] [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: 04/22/2024] [Accepted: 07/31/2024] [Indexed: 09/12/2024] Open
Abstract
The conventional real-time screening in organs-on-chips is limited to optical tracking of pre-tagged cells and biological agents. This work introduces an efficient biofabrication protocol to integrate tunable hydrogel electrodes into 3D bioprinted-on-chips. We established our method of fabricating cell-laden hydrogel-based microfluidic chips through digital light processing-based 3D bioprinting. Our conductive ink includes poly-(3,4-ethylene-dioxythiophene)-polystyrene sulfonate (PEDOT: PSS) microparticles doped in polyethylene glycol diacrylate (PEGDA). We optimized the manufacturing process of PEDOT: PSS microparticles characterized our conductive ink for different 3D bioprinting parameters, geometries, and materials conditions. While the literature is limited to 0.5% w/v for PEDOT: PSS microparticle concentration, we increased their concentration to 5% w/v with superior biological responses. We measured the conductivity in the 3-15 m/m for a range of 0.5%-5% w/v microparticles, and we showed the effectiveness of 3D-printed electrodes for predicting cell responses when encapsulated in gelatin-methacryloyl (GelMA). Interestingly, a higher cellular activity was observed in the case of 5% w/v microparticles compared to 0.5% w/v microparticles. Electrochemical impedance spectroscopy measurements indicated significant differences in cell densities and spheroid sizes embedded in GelMA microtissues.
Collapse
Affiliation(s)
- Ayda Pourmostafa
- Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Anant Bhusal
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ, United States
| | - Niranjan Haridas Menon
- Department of Chemical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Glassboro, Newark, NJ, United States
| | - Zhenglong Li
- Department of Chemical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Glassboro, Newark, NJ, United States
| | - Sagnik Basuray
- Department of Chemical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Glassboro, Newark, NJ, United States
| | - Amir K Miri
- Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| |
Collapse
|
24
|
Wei K, Tang C, Ma H, Fang X, Yang R. 3D-printed microrobots for biomedical applications. Biomater Sci 2024; 12:4301-4334. [PMID: 39041236 DOI: 10.1039/d4bm00674g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Microrobots, which can perform tasks in difficult-to-reach parts of the human body under their own or external power supply, are potential tools for biomedical applications, such as drug delivery, microsurgery, imaging and monitoring, tissue engineering, and sensors and actuators. Compared with traditional fabrication methods for microrobots, recent improvements in 3D printers enable them to print high-precision microrobots, breaking through the limitations of traditional micromanufacturing technologies that require high skills for operators and greatly shortening the design-to-production cycle. Here, this review first introduces typical 3D printing technologies used in microrobot manufacturing. Then, the structures of microrobots with different functions and application scenarios are discussed. Next, we summarize the materials (body materials, propulsion materials and intelligent materials) used in 3D microrobot manufacturing to complete body construction and realize biomedical applications (e.g., drug delivery, imaging and monitoring). Finally, the challenges and future prospects of 3D printed microrobots in biomedical applications are discussed in terms of materials, manufacturing and advancement.
Collapse
Affiliation(s)
- Kun Wei
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Chenlong Tang
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Hui Ma
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Xingmiao Fang
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Runhuai Yang
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
25
|
Jin R, Li Y, Xu Y, Cheng L, Jiang D. Stereoscopic Imaging of Single Molecules at Plasma Membrane of Single Cell Using Photoreduction-Assisted Electrochemistry. RESEARCH (WASHINGTON, D.C.) 2024; 7:0443. [PMID: 39140091 PMCID: PMC11319615 DOI: 10.34133/research.0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/14/2024] [Indexed: 08/15/2024]
Abstract
Stereoscopic imaging of single molecules at the plasma membrane of single cell requires spatial resolutions in 3 dimensions (x-y-z) at 10-nm level, which is rarely achieved using most optical super-resolution microscopies. Here, electrochemical stereoscopic microscopy with a detection limit down to a single molecule is achieved using a photoreduction-assisted cycle inside a 20-nm gel electrolyte nanoball at the tip of a nanopipette. On the basis of the electrochemical oxidation of Ru(bpy)3 2+ into Ru(bpy)3 3+ followed by the reduction of Ru(bpy)3 3+ into Ru(bpy)3 2+ by photogenerated isopropanol radicals, a charge of 1.5 fC is obtained from the cycling electron transfers involving one Ru(bpy)3 2+/3+ molecule. By using the nanopipette to scan the cellular membrane modified with Ru(bpy)3 2+-tagged antibody, the morphology of the cell membrane and the distribution of carcinoembryonic antigen (CEA) on the membrane are electrochemically visualized with a spatial resolution of 14 nm. The resultant stereoscopic image reveals more CEA on membrane protrusions, providing direct evidence to support easy access of membrane CEA to intravenous antibodies. The breakthrough in single-molecule electrochemistry at the cellular level leads to the establishment of high-resolution 3-dimensional single-cell electrochemical microscopy, offering an alternative strategy to remedy the imperfection of stereoscopic visualization in optical microscopes.
Collapse
Affiliation(s)
- Rong Jin
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering,
Nanjing University, Nanjing, China
| | - Yu Li
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering,
Nanjing University, Nanjing, China
| | - Yanyan Xu
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering,
Nanjing University, Nanjing, China
| | - Lei Cheng
- College of Engineering and Technology,
Southwest University, Chongqing, China
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering,
Nanjing University, Nanjing, China
| |
Collapse
|
26
|
O'Shea TC, Croland KJ, Salem A, Urbanski R, Schultz KM. A Rheological Study on the Effect of Tethering Pro- and Anti-Inflammatory Cytokines into Hydrogels on Human Mesenchymal Stem Cell Migration, Degradation, and Morphology. Biomacromolecules 2024; 25:5121-5137. [PMID: 38961715 DOI: 10.1021/acs.biomac.4c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Polymer-peptide hydrogels are being designed as implantable materials that deliver human mesenchymal stem cells (hMSCs) to treat wounds. Most wounds can progress through the healing process without intervention. During the normal healing process, cytokines are released from the wound to create a concentration gradient, which causes directed cell migration from the native niche to the wound site. Our work takes inspiration from this process and uniformly tethers cytokines into the scaffold to measure changes in cell-mediated degradation and motility. This is the first step in designing cytokine concentration gradients into the material to direct cell migration. We measure changes in rheological properties, encapsulated cell-mediated pericellular degradation and migration in a hydrogel scaffold with covalently tethered cytokines, either tumor necrosis factor-α (TNF-α) or transforming growth factor-β (TGF-β). TNF-α is expressed in early stages of wound healing causing an inflammatory response. TGF-β is released in later stages of wound healing causing an anti-inflammatory response in the surrounding tissue. Both cytokines cause directed cell migration. We measure no statistically significant difference in modulus or the critical relaxation exponent when tethering either cytokine in the polymeric network without encapsulated hMSCs. This indicates that the scaffold structure and rheology is unchanged by the addition of tethered cytokines. Increases in hMSC motility, morphology and cell-mediated degradation are measured using a combination of multiple particle tracking microrheology (MPT) and live-cell imaging in hydrogels with tethered cytokines. We measure that tethering TNF-α into the hydrogel increases cellular remodeling on earlier days postencapsulation and tethering TGF-β into the scaffold increases cellular remodeling on later days. We measure tethering either TGF-β or TNF-α enhances cell stretching and, subsequently, migration. This work provides rheological characterization that can be used to design new materials that present chemical cues in the pericellular region to direct cell migration.
Collapse
Affiliation(s)
- Thomas C O'Shea
- Purdue University, Davidson School of Chemical Engineering, 480 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Kiera J Croland
- University of Colorado at Boulder, Department of Chemical and Biological Engineering, 3415 Colorado Ave, Boulder, Colorado 80303, United States
| | - Ahmad Salem
- Lehigh University, Department of Chemical and Biomolecular Engineering, 124 East Morton Street, Bethlehem, Pennsylvania 18015, United States
| | - Rylie Urbanski
- Lehigh University, Department of Chemical and Biomolecular Engineering, 124 East Morton Street, Bethlehem, Pennsylvania 18015, United States
| | - Kelly M Schultz
- Purdue University, Davidson School of Chemical Engineering, 480 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
27
|
Cianciosi A, Pfeiffle M, Wohlfahrt P, Nürnberger S, Jungst T. Optical Fiber-Assisted Printing: A Platform Technology for Straightforward Photopolymer Resins Patterning and Freeform 3D Printing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403049. [PMID: 39189485 PMCID: PMC11348141 DOI: 10.1002/advs.202403049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/31/2024] [Indexed: 08/28/2024]
Abstract
Light-based 3D printing techniques represent powerful tools, enabling the precise fabrication of intricate objects with high resolution and control. An innovative addition to this set of printing techniques is Optical Fiber-Assisted Printing (OFAP) introduced in this article. OFAP is a platform utilizing an LED-coupled optical fiber (LOF) that selectively crosslinks photopolymer resins. It allows change of parameters like light intensity and LOF velocity during fabrication, facilitating the creation of structures with progressive features and multi-material constructs layer-by-layer. An optimized formulation based on allyl-modified gelatin (gelAGE) with food dyes as photoabsorbers is introduced. Additionally, a novel gelatin-based biomaterial, alkyne-modified gelatin (gelGPE), featuring alkyne moieties, demonstrates near-visible light absorption thus fitting OFAP needs, paving the way for multifunctional hydrogels through thiol-yne click chemistry. Besides 2D patterning, OFAP is transferred to embedded 3D printing within a resin bath demonstrating the proof-of-concept as a novel printing technology with potential applications in tissue engineering and biomimetic scaffold fabrication, offering rapid and precise freeform printing capabilities.
Collapse
Affiliation(s)
- Alessandro Cianciosi
- Department for Functional Materials in Medicine and DentistryInstitute of Functional Materials and BiofabricationUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Maximilian Pfeiffle
- Department for Functional Materials in Medicine and DentistryInstitute of Functional Materials and BiofabricationUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Philipp Wohlfahrt
- Department for Functional Materials in Medicine and DentistryInstitute of Functional Materials and BiofabricationUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Severin Nürnberger
- Department for Functional Materials in Medicine and DentistryInstitute of Functional Materials and BiofabricationUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| | - Tomasz Jungst
- Department for Functional Materials in Medicine and DentistryInstitute of Functional Materials and BiofabricationUniversity of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI)Pleicherwall 297070WürzburgGermany
| |
Collapse
|
28
|
Salisbury E, Rawlings TM, Efstathiou S, Tryfonos M, Makwana K, Fitzgerald HC, Gargett CE, Cameron NR, Haddleton DM, Brosens JJ, Eissa AM. Photo-Cross-linked Gelatin Methacryloyl Hydrogels Enable the Growth of Primary Human Endometrial Stromal Cells and Epithelial Gland Organoids. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39140-39152. [PMID: 39022819 PMCID: PMC11299152 DOI: 10.1021/acsami.4c08763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
In vitro three-dimensional (3D) models are better able to replicate the complexity of real organs and tissues than 2D monolayer models. The human endometrium, the inner lining of the uterus, undergoes complex changes during the menstrual cycle and pregnancy. These changes occur in response to steroid hormone fluctuations and elicit crosstalk between the epithelial and stromal cell compartments, and dysregulations are associated with a variety of pregnancy disorders. Despite the importance of the endometrium in embryo implantation and pregnancy establishment, there is a lack of in vitro models that recapitulate tissue structure and function and as such a growing demand for extracellular matrix hydrogels that can support 3D cell culture. To be physiologically relevant, an in vitro model requires mechanical and biochemical cues that mimic those of the ECM found in the native tissue. We report a semisynthetic gelatin methacryloyl (GelMA) hydrogel that combines the bioactive properties of natural hydrogels with the tunability and reproducibility of synthetic materials. We then describe a simple protocol whereby cells can quickly be encapsulated in GelMA hydrogels. We investigate the suitability of GelMA hydrogel to support the development of an endometrial model by culturing the main endometrial cell types: stromal cells and epithelial cells. We also demonstrate how the mechanical and biochemical properties of GelMA hydrogels can be tailored to support the growth and maintenance of epithelial gland organoids that emerge upon 3D culturing of primary endometrial epithelial progenitor cells in a defined chemical medium. We furthermore demonstrate the ability of GelMA hydrogels to support the viability of stromal cells and their function measured by monitoring decidualization in response to steroid hormones. This study describes the first steps toward the development of a hydrogel matrix-based model that recapitulates the structure and function of the native endometrium and could support applications in understanding reproductive failure.
Collapse
Affiliation(s)
- Emma Salisbury
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Thomas M. Rawlings
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | | | - Maria Tryfonos
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | - Komal Makwana
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | - Harriet C. Fitzgerald
- The
Ritchie Centre, Hudson Institute of Medical Research, Clayton VIC 3168, Australia
- Department
of Obstetrics and Gynaecology, Monash University, Clayton VIC 3168, Australia
| | - Caroline E. Gargett
- The
Ritchie Centre, Hudson Institute of Medical Research, Clayton VIC 3168, Australia
- Department
of Obstetrics and Gynaecology, Monash University, Clayton VIC 3168, Australia
| | - Neil R. Cameron
- Department
of Materials Science and Engineering, Monash
University, Clayton, Victoria 3800, Australia
- School of
Engineering, University of Warwick, Coventry CV4 7AL, U.K.
| | | | - Jan J. Brosens
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | - Ahmed M. Eissa
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Polymers, Chemical Industries Research Division, National Research
Centre, El Bohouth St.
33, Dokki, Cairo Giza 12622, Egypt
- School
of Life Sciences, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, U.K.
| |
Collapse
|
29
|
Chang CW, Dargaville BL, Momot KI, Hutmacher DW. An investigation of water status in gelatin methacrylate hydrogels by means of water relaxometry and differential scanning calorimetry. J Mater Chem B 2024; 12:6328-6341. [PMID: 38628083 DOI: 10.1039/d4tb00053f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The relationship between molecular structure and water dynamics is a fundamental yet often neglected subject in the field of hydrogels for drug delivery, bioprinting, as well as biomaterial science and tissue engineering & regenerative medicine (TE&RM). Water is a fundamental constituent of hydrogel systems and engages via hydrogen bonding with the macromolecular network. The methods and techniques to measure and reveal the phenomena and dynamics of water within hydrogels are still limited. In this work, differential scanning calorimetry (DSC) was used as a quantitative method to analyze freezable (including free and freezable bound) and non-freezable bound water within gelatin methacrylate (GelMA) hydrogels. Nuclear magnetic resonance (NMR) is a complementary method for the study of water behavior and can be used to measure the spin-relaxation of water hydrogen nuclei, which is related to water dynamics. In this research, nuclear magnetic resonance relaxometry was employed to investigate the molecular state of water in GelMA hydrogels using spin-lattice (T1) and spin-spin (T2) spin-relaxation time constants. The data displays a trend of increasing bound water content with increasing GelMA concentration. In addition, T2 values were further applied to calculate microviscosity and translational diffusion coefficients. Water relaxation under various chemical environments, including different media, temperatures, gelatin sources, as well as crosslinking effects, were also examined. These comprehensive physical data sets offer fundamental insight into biomolecule transport within the GelMA hydrogel system, which ultimately are important for drug delivery, bioprinting, as well as biomaterial science and TE&RM communities.
Collapse
Affiliation(s)
- Chun-Wei Chang
- Max Planck Queensland Centre on the Materials Science for Extracellular Matrices, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), QLD 4059, Australia
| | - Bronwin L Dargaville
- Max Planck Queensland Centre on the Materials Science for Extracellular Matrices, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), QLD 4059, Australia
| | - Konstantin I Momot
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Dietmar W Hutmacher
- Max Planck Queensland Centre on the Materials Science for Extracellular Matrices, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), QLD 4059, Australia
| |
Collapse
|
30
|
Zatorski JM, Lee IL, Ortiz-Cárdenas JE, Ellena JF, Pompano RR. Measurement of covalent bond formation in light-curing hydrogels predicts physical stability under flow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.30.601353. [PMID: 39005331 PMCID: PMC11244878 DOI: 10.1101/2024.06.30.601353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Photocrosslinking hydrogels are promising for tissue engineering and regenerative medicine, but challenges in reaction monitoring often leave their optimization subject to trial and error. The stability of crosslinked gels under fluid flow, as in the case of a microfluidic device, is particularly challenging to predict, both because of obstacles inherent to solid-state macromolecular analysis that prevent accurate chemical monitoring, and because stability is dependent on size of the patterned features. To solve both problems, we obtained 1H NMR spectra of cured hydrogels which were enzymatically degraded. This allowed us to take advantage of the high-resolution that solution NMR provides. This unique approach enabled the measurement of degree of crosslinking (DoC) and prediction of material stability under physiological fluid flow. We showed that NMR spectra of enzyme-digested gels successfully reported on DoC as a function of light exposure and wavelength within two classes of photocrosslinkable hydrogels: methacryloyl-modified gelatin and a composite of thiol-modified gelatin and norbornene-terminated polyethylene glycol. This approach revealed that a threshold DoC was required for patterned features in each material to become stable, and that smaller features required a higher DoC for stability. Finally, we demonstrated that DoC was predictive of the stability of architecturally complex features when photopatterning, underscoring the value of monitoring DoC when using light-reactive gels. We anticipate that the ability to quantify chemical crosslinks will accelerate the design of advanced hydrogel materials for structurally demanding applications such as photopatterning and bioprinting.
Collapse
Affiliation(s)
- Jonathan M Zatorski
- University of Virginia, Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Isabella L Lee
- University of Virginia, Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Jennifer E Ortiz-Cárdenas
- Stanford University, Department of Bioengineering, 443 Via Ortega, Rm 119, Stanford, CA 94305, United States
| | - Jeffrey F Ellena
- University of Virginia, Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Rebecca R Pompano
- University of Virginia, Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Sciences, Thornton Hall, 351 McCormick Rd, Charlottesville, VA 22904
- Carter Immunology Center and UVA Cancer Center, University of Virginia, 345 Crispell Dr., MR-6, Charlottesville, VA 22908
| |
Collapse
|
31
|
Zhou G, Li T, Du J, Wu M, Lin D, Pu W, Zhang J, Gu Z. Harnessing HetHydrogel: A Universal Platform to Dropletize Single-Cell Multiomics. SMALL METHODS 2024; 8:e2301631. [PMID: 38419597 DOI: 10.1002/smtd.202301631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/12/2024] [Indexed: 03/02/2024]
Abstract
A universal platform is developed for dropletizing single cell plate-based multiomic assays, consisting of three main pillars: a miniaturized open Heterogeneous Hydrogel reactor (abbreviated HetHydrogel) for multi-step biochemistry, its tunable permeability that allows Tn5 tagmentation, and single cell droplet barcoding. Through optimizing the HetHydrogel manufacturing procedure, the chemical composition, and cell permeation conditions, simultaneous high-throughput mitochondrial DNA genotyping and chromatin profiling at the single-cell level are demonstrated using a mixed-species experiment. This platform offers a powerful way to investigate the genotype-phenotype relationships of various mtDNA mutations in biological processes. The HetHydrogel platform is believed to have the potential to democratize droplet technologies, upgrading a whole range of plate-based single cell assays to high throughput format.
Collapse
Affiliation(s)
- Guoqiang Zhou
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Ting Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Jingjing Du
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Mengying Wu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Deng Lin
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Weilin Pu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Jingwei Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
- Zhejiang Lab, Hangzhou, 310000, China
| | - Zhenglong Gu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| |
Collapse
|
32
|
Hebner TS, Kirkpatrick BE, Fairbanks BD, Bowman CN, Anseth KS, Benoit DS. Radical-Mediated Degradation of Thiol-Maleimide Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402191. [PMID: 38582514 PMCID: PMC11220706 DOI: 10.1002/advs.202402191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/22/2024] [Indexed: 04/08/2024]
Abstract
Michael addition between thiol- and maleimide-functionalized molecules is a long-standing approach used for bioconjugation, hydrogel crosslinking, and the functionalization of other advanced materials. While the simplicity of this chemistry enables facile synthesis of hydrogels, network degradation is also desirable in many instances. Here, the susceptibility of thiol-maleimide bonds to radical-mediated degradation is reported. Irreversible degradation in crosslinked materials is demonstrated using photoinitiated and chemically initiated radicals in hydrogels and linear polymers. The extent of degradation is shown to be dependent on initiator concentration. Using a model linear polymer system, the radical-mediated mechanism of degradation is elucidated, in which the thiosuccinimide crosslink is converted to a succinimide and a new thioether formed with an initiator fragment. Using laser stereolithography, high-fidelity spatiotemporal control over degradation in crosslinked gels is demonstrated. Ultimately, this work establishes a platform for controllable, radical-mediated degradation in thiol-maleimide hydrogels, further expanding their versatility as functional materials.
Collapse
Affiliation(s)
- Tayler S. Hebner
- Department of BioengineeringUniversity of Oregon6231 University of OregonEugeneOR97403USA
| | - Bruce E. Kirkpatrick
- Department of Chemical and Biological EngineeringUniversity of Colorado Boulder596 UCBBoulderCO80309USA
- BioFrontiers InstituteUniversity of Colorado Boulder596 UCBBoulderCO80309USA
- BioFrontiers Institute Medical Scientist Training ProgramUniversity of Colorado Anschutz Medical Campus13001 East 17th PlaceAuroraCO80045USA
| | - Benjamin D. Fairbanks
- Department of Chemical and Biological EngineeringUniversity of Colorado Boulder596 UCBBoulderCO80309USA
| | - Christopher N. Bowman
- Department of Chemical and Biological EngineeringUniversity of Colorado Boulder596 UCBBoulderCO80309USA
| | - Kristi S. Anseth
- Department of Chemical and Biological EngineeringUniversity of Colorado Boulder596 UCBBoulderCO80309USA
- BioFrontiers InstituteUniversity of Colorado Boulder596 UCBBoulderCO80309USA
| | - Danielle S.W. Benoit
- Department of BioengineeringUniversity of Oregon6231 University of OregonEugeneOR97403USA
| |
Collapse
|
33
|
Li L, Liu H, Zhao Y, Liu X, Dong Y, Luo J, Jiang X, Zhang Y, Zhu Q, Yuan X, Pei X, Zhang L, Li B, Yang S, Gou M. 3D Printing of Maturable Tissue Constructs Using a Cell‐Adaptable Nanocolloidal Hydrogel. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202402341] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Indexed: 01/06/2025]
Abstract
Abstract3D‐printed cell‐laden hydrogels as tissue constructs show great promise in generating living tissues for medicine. Currently, the maturation of 3D‐printed constructs into living tissues remains challenge, since commonly used hydrogels struggle to provide an ideal microenvironment for the seeded cells. In this study, a cell‐adaptable nanocolloidal hydrogel is created for 3D printing of maturable tissue constructs. The nanocolloidal hydrogel is composed of interconnected nanoparticles, which is prepared by the self‐assembly and subsequent photocrosslinking of the gelatin methacryloyl solutions. Cells can get enough space to grow and migrate within the hydrogel through squeezing the flexible nanocolloidal networks. Meanwhile, the nanostructure can promote the seeded cells to proliferate and produce matrix proteins through mechanotransduction. Using digital light process‐based 3D printing technology, it can rapidly customize cartilage tissue constructs. After implantation, these tissue constructs efficiently matured into cartilage tissues for the articular cartilage defect repair and ear cartilage reconstruction in vivo. The 3D printing of maturable tissue constructs using the nanocolloidal hydrogel shows potential for future clinical applications.
Collapse
Affiliation(s)
- Li Li
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Haofan Liu
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Yongchao Zhao
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Xuan Liu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces College of Pharmacy Hunan University of Chinese Medicine Changsha 410208 P. R. China
| | - Yinchu Dong
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Jing Luo
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Xuebing Jiang
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Yi Zhang
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Qi Zhu
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Xin Yuan
- Department of Plastic and Burn Surgery West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Xuan Pei
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Li Zhang
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Boya Li
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Shuai Yang
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Maling Gou
- Department of Biotherapy Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| |
Collapse
|
34
|
Mohammad Mehdipour N, Rajeev A, Kumar H, Kim K, Shor RJ, Natale G. Anisotropic hydrogel scaffold by flow-induced stereolithography 3D printing technique. BIOMATERIALS ADVANCES 2024; 161:213885. [PMID: 38743993 DOI: 10.1016/j.bioadv.2024.213885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
Essential organs, such as the heart and liver, contain a unique porous network that allows oxygen and nutrients to be exchanged, with distinct random to ordered regions displaying varying degrees of strength. A novel technique, referred to here as flow-induced lithography, was developed. This technique generates tunable anisotropic three-dimensional (3D) structures. The ink for this bioprinting technique was made of titanium dioxide nanorods (Ti) and kaolinite nanoclay (KLT) dispersed in a GelMA/PEGDA polymeric suspension. By controlling the flow rate, aligned particle microstructures were achieved in the suspensions. The application of UV light to trigger the polymerization of the photoactive prepolymer freezes the oriented particles in the polymer network. Because the viability test was successful in shearing suspensions containing cells, the flow-induced lithography technique can be used with both acellular scaffolds and cell-laden structures. Fabricated hydrogels show outstanding mechanical properties resembling human tissues, as well as significant cell viability (> 95 %) over one week. As a result of this technique and the introduction of bio-ink, a novel approach has been pioneered for developing anisotropic tissue implants utilizing low-viscosity biomaterials.
Collapse
Affiliation(s)
- Narges Mohammad Mehdipour
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Ashna Rajeev
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Hitendra Kumar
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453552, India
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Roman J Shor
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Giovanniantonio Natale
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
35
|
Almalla A, Alzain N, Elomaa L, Richter F, Scholz J, Lindner M, Siegmund B, Weinhart M. Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models. Cells 2024; 13:1080. [PMID: 38994934 PMCID: PMC11240340 DOI: 10.3390/cells13131080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.
Collapse
Affiliation(s)
- Ahed Almalla
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Nadra Alzain
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Fiona Richter
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Johanna Scholz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Marcus Lindner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstr. 3A, 30167 Hannover, Germany
| |
Collapse
|
36
|
Abraham S, Gupta P, Govarthanan K, Rao S, Santra TS. Direction-oriented fiber guiding with a tunable tri-layer-3D scaffold for periodontal regeneration. RSC Adv 2024; 14:19806-19822. [PMID: 38899033 PMCID: PMC11186324 DOI: 10.1039/d4ra01459f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Multilayered scaffolds mimicking mechanical and biological host tissue architectures are the current prerequisites for successful tissue regeneration. We propose our tunable tri-layered scaffold, designed to represent the native periodontium for potential regenerative applications. The fused deposition modeling platform is used to fabricate the novel movable three-layered polylactic acid scaffold mimicking in vivo cementum, periodontal ligament, and alveolar bone layers. The scaffold is further provided with multiple angulated fibers, offering directional guidance and facilitating the anchorage dependence on cell adhesion. Additionally, surface modifications of the scaffold were made by incorporating coatings like collagen and different concentrations of gelatin methacryloyl to enrich the cell adhesion and proliferation. The surface characterization of our designed scaffolds was performed using tribological studies, atomic force microscopy, contact angle measurement, scanning electron microscopy, and micro-computed tomography. Furthermore, the material characterization of this scaffold was investigated by attenuated total reflectance-Fourier transformed infrared spectroscopy. The scaffold's mechanical characterization, such as strength and compression modulus, was demonstrated by compression testing. The L929 mouse fibroblast cells and MG63 human osteosarcoma cells have been cultured on the scaffold. The scaffold's superior biocompatibility was evaluated using fluorescence dye with fluorescence microscopy, scanning electron microscopy, in vitro wound healing assay, MTT assay, and flow cytometry. The mineralization capability of the scaffolds was also studied. In conclusion, our study demonstrated the construction of a multilayered movable scaffold, which is highly biocompatible and most suitable for various downstream applications such as periodontium and in situ tissue regeneration of complex, multilayered tissues.
Collapse
Affiliation(s)
- Sarin Abraham
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| | - Kavitha Govarthanan
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem) Bengaluru Karnataka 560065 India
| | - Suresh Rao
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| |
Collapse
|
37
|
Abaci A, Guvendiren M. 3D bioprinting of dense cellular structures within hydrogels with spatially controlled heterogeneity. Biofabrication 2024; 16:035027. [PMID: 38821144 DOI: 10.1088/1758-5090/ad52f1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/31/2024] [Indexed: 06/02/2024]
Abstract
Embedded bioprinting is an emerging technology for precise deposition of cell-laden or cell-only bioinks to construct tissue like structures. Bioink is extruded or transferred into a yield stress hydrogel or a microgel support bath allowing print needle motion during printing and providing temporal support for the printed construct. Although this technology has enabled creation of complex tissue structures, it remains a challenge to develop a support bath with user-defined extracellular mimetic cues and their spatial and temporal control. This is crucial to mimic the dynamic nature of the native tissue to better regenerate tissues and organs. To address this, we present a bioprinting approach involving printing of a photocurable viscous support layer and bioprinting of a cell-only or cell-laden bioink within this viscous layer followed by brief exposure to light to partially crosslink the support layer. This approach does not require shear thinning behavior and is suitable for a wide range of photocurable hydrogels to be used as a support. It enables multi-material printing to spatially control support hydrogel heterogeneity including temporal delivery of bioactive cues (e.g. growth factors), and precise patterning of dense multi-cellular structures within these hydrogel supports. Here, dense stem cell aggregates are printed within methacrylated hyaluronic acid-based hydrogels with patterned heterogeneity to spatially modulate human mesenchymal stem cell osteogenesis. This study has significant impactions on creating tissue interfaces (e.g. osteochondral tissue) in which spatial control of extracellular matrix properties for patterned stem cell differentiation is crucial.
Collapse
Affiliation(s)
- Alperen Abaci
- Otto H. York Chemical and Materials Engineering Department, New Jersey Institute of Technology, Newark, NJ, United States of America
| | - Murat Guvendiren
- Otto H. York Chemical and Materials Engineering Department, New Jersey Institute of Technology, Newark, NJ, United States of America
- Bioengineering Department, New Jersey Institute of Technology, Newark, NJ, United States of America
| |
Collapse
|
38
|
Iseki T, Rothrauff BB, Kihara S, Overholt KJ, Taha T, Lin H, Alexander PG, Tuan RS. Enhanced osteochondral repair by leukocyte-depleted platelet-rich plasma in combination with adipose-derived mesenchymal stromal cells encapsulated in a three-dimensional photocrosslinked injectable hydrogel in a rabbit model. Stem Cell Res Ther 2024; 15:159. [PMID: 38831361 PMCID: PMC11149275 DOI: 10.1186/s13287-024-03750-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
INTRODUCTION Intra-articular injection of adipose-derived mesenchymal stromal cells (ASCs) and/or platelet-rich plasma (PRP) have been reported to independently and synergistically improve healing of osteochondral lesions in animal models. However, their independent and combined effects when localized to an osteochondral lesion by encapsulation within a photocrosslinkable methacrylated gelatin hydrogel (GelMA) have not been explored. Herein we investigated a unique combination of allogeneic ASCs and PRP embedded in GelMA as a single-stage treatment for osteochondral regeneration in a rabbit model. METHODS Thirty mature rabbits were divided into six experimental groups: (1) Sham; (2) Defect; (3) GelMA; (4) GelMA + ASCs; (5) GelMA + PRP; and (6) GelMA + ASCs + PRP.At 12 weeks following surgical repair, osteochondral regeneration was assessed on the basis of gross appearance, biomechanical properties, histological and immunohistochemical characteristics, and subchondral bone volume. RESULTS In terms of mechanical property reflecting the ability of neotissue to bear stress, PRP only group were significantly lower than the Sham group (p = 0.0098). On the other hand, ASCs only and ASCs combined with PRP groups did not exhibit significantly difference, which suggesting that incorporation of ASCs assists in restoring the ability of the neotissue to bear stresses similarly to native tissue (p = 0.346, p = 0.40, respectively). Safranin O in ASCs combined with PRP group was significantly higher than the Defect and GelMA only groups (p = 0.0009, p = 0.0017, respectively). Additionally, ASCs only and ASCs combined with PRP groups presented especially strong staining for collagen type II. Surprisingly, PRP only and PRP + ASCs groups tended to exhibit higher collagen type I and collagen type X staining compared to ASCs only group, suggesting a potential PRP-mediated hypertrophic effect. CONCLUSION Regeneration of a focal osteochondral defect in a rabbit model was improved by a single-stage treatment of a photocrosslinked hydrogel containing allogenic ASCs and autologous PRP, with the combination of ASCs and PRP producing superior benefit than either alone. No experimental construct fully restored all properties of the native, healthy osteochondral unit, which may require longer follow-up or further modification of PRP and/or ASCs characteristics.
Collapse
Affiliation(s)
- Tomoya Iseki
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- Department of Orthopaedic Surgery, Hyogo Medical University, 1-1 Mukogawa-cho, 663-8501, Nishinomiya City, Hyogo, Japan
| | - Benjamin B Rothrauff
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Shinsuke Kihara
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Kalon J Overholt
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Tarek Taha
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
39
|
Mazari‐Arrighi E, Lépine M, Ayollo D, Faivre L, Larghero J, Chatelain F, Fuchs A. Self-Organization of Long-Lasting Human Endothelial Capillary-Like Networks Guided by DLP Bioprinting. Adv Healthc Mater 2024; 13:e2302830. [PMID: 38366136 PMCID: PMC11468676 DOI: 10.1002/adhm.202302830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/29/2024] [Indexed: 02/18/2024]
Abstract
Tissue engineering holds great promise for regenerative medicine, drug discovery, and as an alternative to animal models. However, as soon as the dimensions of engineered tissue exceed the diffusion limit of oxygen and nutriments, a necrotic core forms leading to irreversible damage. To overcome this constraint, the establishment of a functional perfusion network is essential. In this work, digital light processing bioprinting is used to encapsulate endothelial progenitor cells (EPCs) in 3D light-cured hydrogel scaffolds to guide them toward vascular network formation. In these scaffolds, EPCs proliferate and self-organize within a few days into branched tubular structures with predefined geometry, forming capillary-like vascular tubes or trees of diameters in the range of 10 to 100 µm. Presenting a confluent monolayer wall of cells strongly connect by tight junctions around a central lumen-like space, these structures can be microinjected with a fluorescent dye and are stable for several weeks in vitro. These endothelial structures can be recovered and manipulated in an alginate patch without altering their shape or viability. This approach opens new opportunities for future applications, such as stacking with other cell sheets or multicellular constructs to yield bioengineered tissue with higher complexity and functionality.
Collapse
Affiliation(s)
- Elsa Mazari‐Arrighi
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Matthieu Lépine
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Dmitry Ayollo
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Lionel Faivre
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - Jérôme Larghero
- Université de ParisU976 HIPI, InsermParisF‐75006France
- AP‐HPHôpital Saint‐Louis1 avenue VellefauxParisF‐75010France
| | - François Chatelain
- Université de ParisU976 HIPI, InsermParisF‐75006France
- CEAIRIGGrenobleF‐38000France
| | - Alexandra Fuchs
- Université de ParisU976 HIPI, InsermParisF‐75006France
- CEAIRIGGrenobleF‐38000France
| |
Collapse
|
40
|
Li X, Li Y, Zhang X, Xu J, Kang J, Li B, Zhao B, Wang L. Cross-Linking Methods of the Silk Protein Hydrogel in Oral and Craniomaxillofacial Tissue Regeneration. Tissue Eng Regen Med 2024; 21:529-544. [PMID: 38294593 PMCID: PMC11087422 DOI: 10.1007/s13770-023-00624-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Craniomaxillofacial tissue defects are clinical defects involving craniomaxillofacial and oral soft and hard tissues. They are characterized by defect-shaped irregularities, bacterial and inflammatory environments, and the need for functional recovery. Conventional clinical treatments are currently unable to achieve regeneration of high-quality oral craniomaxillofacial tissue. As a natural biomaterial, silk fibroin (SF) has been widely studied in biomedicine and has broad prospects for use in tissue regeneration. Hydrogels made of SF showed excellent water retention, biocompatibility, safety and the ability to combine with other materials. METHODS To gain an in-depth understanding of the current development of SF, this article reviews the structure, preparation and application prospects in oral and craniomaxillofacial tissue regenerative medicine. It first briefly introduces the structure of SF and then summarizes the principles, advantages and disadvantages of the different cross-linking methods (physical cross-linking, chemical cross-linking and double network structure) of SF. Finally, the existing research on the use of SF in tissue engineering and the prospects of using SF with different cross-linking methods in oral and craniomaxillofacial tissue regeneration are also discussed. CONCLUSIONS This review is intended to show the advantages of SF hydrogels in tissue engineering and provides theoretical support for establishing novel and viable silk protein hydrogels for regeneration.
Collapse
Affiliation(s)
- Xiujuan Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Yuanjiao Li
- School of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinsong Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Jie Xu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Jie Kang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Bing Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Bin Zhao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
| | - Lu Wang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
41
|
Mahajan A, Zaidi ZS, Shukla A, Saxena R, Katti DS. Functionally graded hydrogels with opposing biochemical cues for osteochondral tissue engineering. Biofabrication 2024; 16:035020. [PMID: 38697073 DOI: 10.1088/1758-5090/ad467e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Osteochondral tissue (OC) repair remains a significant challenge in the field of musculoskeletal tissue engineering. OC tissue displays a gradient structure characterized by variations in both cell types and extracellular matrix components, from cartilage to the subchondral bone. These functional gradients observed in the native tissue have been replicated to engineer OC tissuein vitro. While diverse fabrication methods have been employed to create these microenvironments, emulating the natural gradients and effective regeneration of the tissue continues to present a significant challenge. In this study, we present the design and development of CMC-silk interpenetrating (IPN) hydrogel with opposing dual biochemical gradients similar to native tissue with the aim to regenerate the complete OC unit. The gradients of biochemical cues were generated using an in-house-built extrusion system. Firstly, we fabricated a hydrogel that exhibits a smooth transition of sulfated carboxymethyl cellulose (sCMC) and TGF-β1 (SCT gradient hydrogel) from the upper to the lower region of the IPN hydrogel to regenerate the cartilage layer. Secondly, a hydrogel with a hydroxyapatite (HAp) gradient (HAp gradient hydrogel) from the lower to the upper region was fabricated to facilitate the regeneration of the subchondral bone layer. Subsequently, we developed a dual biochemical gradient hydrogel with a smooth transition of sCMC + TGF-β1 and HAp gradients in opposing directions, along with a blend of both biochemical cues in the middle. The results showed that the dual biochemical gradient hydrogels with biochemical cues corresponding to the three zones (i.e. cartilage, interface and bone) of the OC tissue led to differentiation of bone-marrow-derived mesenchymal stem cells to zone-specific lineages, thereby demonstrating their efficacy in directing the fate of progenitor cells. In summary, our study provided a simple and innovative method for incorporating gradients of biochemical cues into hydrogels. The gradients of biochemical cues spatially guided the differentiation of stem cells and facilitated tissue growth, which would eventually lead to the regeneration of the entire OC tissue with a smooth transition from cartilage (soft) to bone (hard) tissues. This promising approach is translatable and has the potential to generate numerous biochemical and biophysical gradients for regeneration of other interface tissues, such as tendon-to-muscle and ligament-to-bone.
Collapse
Affiliation(s)
- Aman Mahajan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Zahra Sifat Zaidi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Amit Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Rakshita Saxena
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology-Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
42
|
Kunwar P, Aryal U, Poudel A, Fougnier D, Geffert ZJ, Xie R, Li Z, Soman P. Droplet bioprinting of acellular and cell-laden structures at high-resolutions. Biofabrication 2024; 16:035019. [PMID: 38749419 DOI: 10.1088/1758-5090/ad4c09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/15/2024] [Indexed: 05/24/2024]
Abstract
Advances in digital light projection(DLP) based (bio) printers have made printing of intricate structures at high resolution possible using a wide range of photosensitive bioinks. A typical setup of a DLP bioprinter includes a vat or reservoir filled with liquid bioink, which presents challenges in terms of cost associated with bioink synthesis, high waste, and gravity-induced cell settling, contaminations, or variation in bioink viscosity during the printing process. Here, we report a vat-free, low-volume, waste-free droplet bioprinting method capable of rapidly printing 3D soft structures at high resolution using model bioinks and model cells. A multiphase many-body dissipative particle dynamics model was developed to simulate the dynamic process of droplet-based DLP printing and elucidate the roles of surface wettability and bioink viscosity. Process variables such as light intensity, photo-initiator concentration, and bioink formulations were optimized to print 3D soft structures (∼0.4-3 kPa) with a typical layer thickness of 50µm, an XY resolution of 38 ± 1.5μm and Z resolution of 237 ± 5.4µm. To demonstrate its versatility, droplet bioprinting was used to print a range of acellular 3D structures such as a lattice cube, a Mayan pyramid, a heart-shaped structure, and a microfluidic chip with endothelialized channels. Droplet bioprinting, performed using model C3H/10T1/2 cells, exhibited high viability (90%) and cell spreading. Additionally, microfluidic devices with internal channel networks lined with endothelial cells showed robust monolayer formation while osteoblast-laden constructs showed mineral deposition upon osteogenic induction. Overall, droplet bioprinting could be a low-cost, no-waste, easy-to-use, method to make customized bioprinted constructs for a range of biomedical applications.
Collapse
Affiliation(s)
- Puskal Kunwar
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Ujjwal Aryal
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Arun Poudel
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Daniel Fougnier
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Zachary J Geffert
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Rui Xie
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| | - Zhen Li
- Department of Mechanical Engineering, Clemson University, Clemson, SC 29634, United States of America
| | - Pranav Soman
- Biomedical, and Chemical Engineering Department, Syracuse University, Syracuse, NY 13210, United States of America
- BioInspired Institute, Syracuse, NY 13210, United States of America
| |
Collapse
|
43
|
Khine YY, Nguyen H, Afolabi F, Lin CC. Fast-relaxing hydrogels with reversibly tunable mechanics for dynamic cancer cell culture. BIOMATERIALS ADVANCES 2024; 159:213829. [PMID: 38531258 PMCID: PMC11075809 DOI: 10.1016/j.bioadv.2024.213829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
The mechanics of the tumor microenvironment (TME) significantly impact disease progression and the efficacy of anti-cancer therapeutics. While it is recognized that advanced in vitro cancer models will benefit cancer research, none of the current engineered extracellular matrices (ECM) adequately recapitulate the highly dynamic TME. Through integrating reversible boronate-ester bonding and dithiolane ring-opening polymerization, we fabricated synthetic polymer hydrogels with tumor-mimetic fast relaxation and reversibly tunable elastic moduli. Importantly, the crosslinking and dynamic stiffening of matrix mechanics were achieved in the absence of a photoinitiator, often the source of cytotoxicity. Central to this strategy was Poly(PEGA-co-LAA-co-AAPBA) (PELA), a highly defined polymer synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization. PELA contains dithiolane for initiator-free gel crosslinking, stiffening, and softening, as well as boronic acid for complexation with diol-containing polymers to give rise to tunable viscoelasticity. PELA hydrogels were highly cytocompatible for dynamic culture of patient-derived pancreatic cancer cells. It was found that the fast-relaxing matrix induced mesenchymal phenotype of cancer cells, and dynamic matrix stiffening restricted tumor spheroid growth. Moreover, this new dynamic viscoelastic hydrogel system permitted sequential stiffening and softening to mimic the physical changes of TME.
Collapse
Affiliation(s)
- Yee Yee Khine
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Han Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Favour Afolabi
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
| |
Collapse
|
44
|
Selvaraj S, Chauhan A, Verma R, Dutta V, Rana G, Duglet R, Subbarayan R, Batoo KM. Role of degrading hydrogels in hepatocellular carcinoma drug delivery applications: A review. J Drug Deliv Sci Technol 2024; 95:105628. [DOI: 10.1016/j.jddst.2024.105628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
45
|
Wang R, He X, Su S, Bai J, Liu H, Zhou F. Multifunctional tannic acid-based nanocomposite methacrylated silk fibroin hydrogel with the ability to scavenge reactive oxygen species and reduce inflammation for bone regeneration. Int J Biol Macromol 2024; 266:131357. [PMID: 38580010 DOI: 10.1016/j.ijbiomac.2024.131357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
The microenvironment of bone defect site is vital for bone regeneration. Severe bone defect is often accompanied with severe inflammation and elevated generation of reactive oxygen species (ROS) during bone repair. In recent years, the unfriendly local microenvironment has been paid more and more attention. Some bioactive materials with the ability to regulate the microenvironment to promote bone regeneration urgently need to be developed. Here, we develop a multifunctional composite hydrogel composed of photo-responsive methacrylate silk fibroin (SFMA), laponite (LAP) nanocomposite and tannic acid (TA), aiming to endow hydrogel with antioxidant, anti-inflammatory and osteogenic induction ability. Characterization results confirmed that the SFMA-LAP@TA hydrogel could significantly improve the mechanical properties of hydrogel. The ROS-Scavenging ability of the hydrogel enabled bone marrow mesenchymal stem cells (BMSCs) to survive against H2O2-induced oxidative stress. In addition, the SFMA-LAP@TA hydrogel effectively decreased the expression of pro-inflammatory factors in RAW264.7. More importantly, the SFMA-LAP@TA hydrogel could enhance the expression of osteogenic markers of BMSCs under inflammatory condition and greatly promote new bone formation in a critical-sized cranial defect model. Above all, the multifunctional hydrogel could effectively promote bone regeneration in vitro and in vivo by scavenging ROS and reducing inflammation, providing a prospective strategy for bone regeneration.
Collapse
Affiliation(s)
- Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Xi He
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shilong Su
- Department of Orthopedics, Peking University Third Hospital, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Jinwu Bai
- Department of Orthopedics, Peking University Third Hospital, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China; Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| |
Collapse
|
46
|
Soliman BG, Longoni A, Major GS, Lindberg GCJ, Choi YS, Zhang YS, Woodfield TBF, Lim KS. Harnessing Macromolecular Chemistry to Design Hydrogel Micro- and Macro-Environments. Macromol Biosci 2024; 24:e2300457. [PMID: 38035637 DOI: 10.1002/mabi.202300457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Indexed: 12/02/2023]
Abstract
Cell encapsulation within three-dimensional hydrogels is a promising approach to mimic tissues. However, true biomimicry of the intricate microenvironment, biophysical and biochemical gradients, and the macroscale hierarchical spatial organizations of native tissues is an unmet challenge within tissue engineering. This review provides an overview of the macromolecular chemistries that have been applied toward the design of cell-friendly hydrogels, as well as their application toward controlling biophysical and biochemical bulk and gradient properties of the microenvironment. Furthermore, biofabrication technologies provide the opportunity to simultaneously replicate macroscale features of native tissues. Biofabrication strategies are reviewed in detail with a particular focus on the compatibility of these strategies with the current macromolecular toolkit described for hydrogel design and the challenges associated with their clinical translation. This review identifies that the convergence of the ever-expanding macromolecular toolkit and technological advancements within the field of biofabrication, along with an improved biological understanding, represents a promising strategy toward the successful tissue regeneration.
Collapse
Affiliation(s)
- Bram G Soliman
- School of Materials Science and Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Alessia Longoni
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3584CX, The Netherlands
| | - Gretel S Major
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Gabriella C J Lindberg
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, 6009, Australia
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02115, USA
| | - Tim B F Woodfield
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Department of Orthopedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, 8011, New Zealand
- School of Medical Sciences, University of Sydney, Sydney, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
47
|
Yaron JR, Gosangi M, Pallod S, Rege K. In situ light-activated materials for skin wound healing and repair: A narrative review. Bioeng Transl Med 2024; 9:e10637. [PMID: 38818119 PMCID: PMC11135152 DOI: 10.1002/btm2.10637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/22/2023] [Accepted: 12/12/2023] [Indexed: 06/01/2024] Open
Abstract
Dermal wounds are a major global health burden made worse by common comorbidities such as diabetes and infection. Appropriate wound closure relies on a highly coordinated series of cellular events, ultimately bridging tissue gaps and regenerating normal physiological structures. Wound dressings are an important component of wound care management, providing a barrier against external insults while preserving the active reparative processes underway within the wound bed. The development of wound dressings with biomaterial constituents has become an attractive design strategy due to the varied functions intrinsic in biological polymers, such as cell instructiveness, growth factor binding, antimicrobial properties, and tissue integration. Using photosensitive agents to generate crosslinked or photopolymerized dressings in situ provides an opportunity to develop dressings rapidly within the wound bed, facilitating robust adhesion to the wound bed for greater barrier protection and adaptation to irregular wound shapes. Despite the popularity of this fabrication approach, relatively few experimental wound dressings have undergone preclinical translation into animal models, limiting the overall integrity of assessing their potential as effective wound dressings. Here, we provide an up-to-date narrative review of reported photoinitiator- and wavelength-guided design strategies for in situ light activation of biomaterial dressings that have been evaluated in preclinical wound healing models.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
- School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State UniversityTempeArizonaUSA
| | - Mallikarjun Gosangi
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
| | - Shubham Pallod
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
- School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State UniversityTempeArizonaUSA
- Chemical Engineering, Arizona State UniversityTempeArizonaUSA
| |
Collapse
|
48
|
Nobus O, Parmentier L, Livens P, Muyshondt P, Szewcyk K, Jacobs C, Verdoodt D, Pieters L, Thijssen Q, Van Durme B, Vral A, Dirckx J, Van Rompaey V, Van Vlierberghe S. The importance of mechanical and biological cues of tympanic membrane grafts to ensure optimal regeneration. BIOMATERIALS ADVANCES 2024; 159:213827. [PMID: 38490018 DOI: 10.1016/j.bioadv.2024.213827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
Chronic suppurative otitis media (CSOM) is often associated with permanent tympanic membrane (TM) perforation and conductive hearing loss. The current clinical gold standard, using autografts and allografts, suffers from several drawbacks. Artificial replacement materials can help to overcome these drawbacks. Therefore, scaffolds fabricated through digital light processing (DLP) were herein created to support TM regeneration. Various UV-curable printing inks, including gelatin methacryloyl (GelMA), gelatin-norbornene-norbornene (GelNBNB) (crosslinked with thiolated gelatin (GelSH)) and alkene-functionalized poly-ε-caprolactone (E-PCL) (crosslinked with pentaerythritol tetrakis(3-mercaptopropionate) (PETA4SH)) were optimized regarding photo-initiator (PI) and photo-absorber (PA) concentrations through viscosity characterization, photo-rheology and the establishment of working curves for DLP. Our material platform enabled the development of constructs with a range of mechanical properties (plateau storage modulus varying between 15 and 119 kPa). Excellent network connectivity for the GelNBNB and E-PCL constructs was demonstrated (gel fractions >95 %) whereas a post-crosslinking step was required for the GelMA constructs. All samples showed excellent biocompatibility (viability >93 % and metabolic activity >88 %). Finally, in vivo and ex vivo assessments, including histology, vibration and deformation responses measured through laser doppler vibrometry and digital image correlation respectively, were performed to investigate the effects of the scaffolds on the anatomical and physiological regeneration of acute TM perforations in rabbits. The data showed that the most efficient healing with the best functional quality was obtained when both mechanical (obtained with the PCL-based resin) and biological (obtained with the gelatin-based resins) material properties were taken into account.
Collapse
Affiliation(s)
- Oriana Nobus
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry (CMaC), Ghent University, 9000 Ghent, Belgium
| | - Laurens Parmentier
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry (CMaC), Ghent University, 9000 Ghent, Belgium
| | - Pieter Livens
- Laboratory of Biomedical Physics (BIMEF), University of Antwerp, 2020 Antwerp, Belgium
| | - Pieter Muyshondt
- Laboratory of Biomedical Physics (BIMEF), University of Antwerp, 2020 Antwerp, Belgium
| | - Krystyna Szewcyk
- Department of Translational Neurosciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Christel Jacobs
- Department of Translational Neurosciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Dorien Verdoodt
- Department of Translational Neurosciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Leen Pieters
- Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
| | - Quinten Thijssen
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry (CMaC), Ghent University, 9000 Ghent, Belgium
| | - Bo Van Durme
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry (CMaC), Ghent University, 9000 Ghent, Belgium
| | - Anne Vral
- Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
| | - Joris Dirckx
- Laboratory of Biomedical Physics (BIMEF), University of Antwerp, 2020 Antwerp, Belgium
| | - Vincent Van Rompaey
- Department of Translational Neurosciences, University of Antwerp, 2610 Wilrijk, Belgium; Department of Otorhinolaryngology and Head & Neck Surgery, Antwerp University Hospital, 2650 Edegem, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry (CMaC), Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
49
|
Klak M, Rachalewski M, Filip A, Dobrzański T, Berman A, Wszoła M. Bioprinting of Perfusable, Biocompatible Vessel-like Channels with dECM-Based Bioinks and Living Cells. Bioengineering (Basel) 2024; 11:439. [PMID: 38790306 PMCID: PMC11117567 DOI: 10.3390/bioengineering11050439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
There is a growing interest in the production of bioinks that on the one hand, are biocompatible and, on the other hand, have mechanical properties that allow for the production of stable constructs that can survive for a long time after transplantation. While the selection of the right material is crucial for bioprinting, there is another equally important issue that is currently being extensively researched-the incorporation of the vascular system into the fabricated scaffolds. Therefore, in the following manuscript, we present the results of research on bioink with unique physico-chemical and biological properties. In this article, two methods of seeding cells were tested using bioink B and seeding after bioprinting the whole model. After 2, 5, 8, or 24 h of incubation, the flow medium was used in the tested systems. At the end of the experimental trial, for each time variant, the canals were stored in formaldehyde, and immunohistochemical staining was performed to examine the presence of cells on the canal walls and roof. Cells adhered to both ways of fiber arrangement; however, a parallel bioprint with the 5 h incubation and the intermediate plating of cells resulted in better adhesion efficiency. For this test variant, the percentage of cells that adhered was at least 20% higher than in the other analyzed variants. In addition, it was for this variant that the lowest percentage of viable cells was found that were washed out of the tested model. Importantly, hematoxylin and eosin staining showed that after 8 days of culture, the cells were evenly distributed throughout the canal roof. Our study clearly shows that neovascularization-promoting cells effectively adhere to ECM-based pancreatic bioink. Summarizing the presented results, it was demonstrated that the proposed bioink compositions can be used for bioprinting bionic organs with a vascular system formed by endothelial cells and fibroblasts.
Collapse
Affiliation(s)
- Marta Klak
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
- Polbionica sp. z o.o., 01-242 Warsaw, Poland
| | - Michał Rachalewski
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
| | - Anna Filip
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
| | | | | | - Michał Wszoła
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
- Polbionica sp. z o.o., 01-242 Warsaw, Poland
| |
Collapse
|
50
|
Isaac AH, Recalde Phillips SY, Ruben E, Estes M, Rajavel V, Baig T, Paleti C, Landsgaard K, Lee RH, Guda T, Criscitiello MF, Gregory C, Alge DL. Impact of PEG sensitization on the efficacy of PEG hydrogel-mediated tissue engineering. Nat Commun 2024; 15:3283. [PMID: 38637507 PMCID: PMC11026400 DOI: 10.1038/s41467-024-46327-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/22/2024] [Indexed: 04/20/2024] Open
Abstract
While poly(ethylene glycol) (PEG) hydrogels are generally regarded as biologically inert blank slates, concerns over PEG immunogenicity are growing, and the implications for tissue engineering are unknown. Here, we investigate these implications by immunizing mice against PEG to stimulate anti-PEG antibody production and evaluating bone defect regeneration after treatment with bone morphogenetic protein-2-loaded PEG hydrogels. Quantitative analysis reveals that PEG sensitization increases bone formation compared to naive controls, whereas histological analysis shows that PEG sensitization induces an abnormally porous bone morphology at the defect site, particularly in males. Furthermore, immune cell recruitment is higher in PEG-sensitized mice administered the PEG-based treatment than their naive counterparts. Interestingly, naive controls that were administered a PEG-based treatment also develop anti-PEG antibodies. Sex differences in bone formation and immune cell recruitment are also apparent. Overall, these findings indicate that anti-PEG immune responses can impact tissue engineering efficacy and highlight the need for further investigation.
Collapse
Affiliation(s)
- Alisa H Isaac
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Elizabeth Ruben
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Matthew Estes
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Varsha Rajavel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Talia Baig
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Carol Paleti
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, College Station, TX, USA
| | - Kirsten Landsgaard
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, College Station, TX, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Michael F Criscitiello
- Comparative Immunogenetics Laboratory, Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | - Carl Gregory
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, College Station, TX, USA
| | - Daniel L Alge
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|