1
|
Fredrikson JP, Roth DM, Cosgrove JA, Sener G, Crow LA, Eckenstein K, Wu L, Hosseini M, Thomas G, Eksi SE, Bertassoni L. Engineering neuronal networks in granular microgels to innervate bioprinted cancer organoids on-a-chip. LAB ON A CHIP 2025. [PMID: 40269972 DOI: 10.1039/d5lc00134j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Organoid models are invaluable for studying organ processes in vitro, offering an unprecedented ability to replicate organ function. Despite recent advancements that have increased their cellular complexity, organoids generally lack key specialized cell types, such as neurons, limiting their ability to fully model organ function and dysfunction. Innervating organoids remains a significant challenge due to the asynchronous biological cues governing neural and organ development. Here, we present a versatile organ-on-a-chip platform designed to innervate organoids across diverse tissue types. Our strategy enables the development of innervated granular hydrogel tissue constructs, followed by the sequential addition of organoids. The microfluidic device features an open tissue chamber, which can be easily manipulated using standard pipetting or advanced bioprinting techniques. Engineered to accommodate microgels of any material larger than 50 μm, the chamber provides flexibility for constructing customizable hydrogel environments. Organoids and other particles can be precisely introduced into the device at any stage using aspiration-assisted bioprinting. To validate this platform, we demonstrate the successful growth of primary mouse superior cervical ganglia (mSCG) neurons and the platform's effectiveness in innervating prostate cancer spheroids and patient-derived renal cell carcinoma organoids. This platform offers a robust and adaptable tool for generating complex innervated organoids, paving the way for more accurate in vitro models of organ development, function, and disease.
Collapse
Affiliation(s)
- Jacob P Fredrikson
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Daniela M Roth
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Jameson A Cosgrove
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Gulsu Sener
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Lily A Crow
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Kazumi Eckenstein
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Lillian Wu
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Mahshid Hosseini
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
| | - George Thomas
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Sebnem Ece Eksi
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Luiz Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| |
Collapse
|
2
|
Kirkpatrick BE, Anseth KS, Hebner TS. Diverse reactivity of maleimides in polymer science and beyond. POLYM INT 2025; 74:296-306. [PMID: 40255264 PMCID: PMC12007691 DOI: 10.1002/pi.6715] [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: 09/16/2024] [Accepted: 10/22/2024] [Indexed: 04/22/2025]
Abstract
Maleimides are remarkably versatile functional groups, capable of participating in homo- and copolymerizations, Diels-Alder and (photo)cycloadditions, Michael additions, and other reactions. Their reactivity has afforded materials ranging from polyimides with high upper service temperatures to hydrogels for regenerative medicine applications. Moreover, maleimides have proven to be an enabling chemistry for pharmaceutical development and bioconjugation via straightforward modification of cysteine residues. To exert spatiotemporal control over reactions with maleimides, multiple approaches have been developed to photocage nucleophiles, dienes, and dipoles. Additionally, further substitution of the maleimide alkene (e.g., mono- and di-halo-, thio-, amino-, and methyl-maleimides, among other substituents) confers tunable reactivity and dynamicity, as well as responsive mechanical and optical properties. In this mini-review, we highlight the diverse functionality of maleimides, underscoring their notable impact in polymer science. This moiety and related heterocycles will play an important role in future innovations in chemistry, biomedical, and materials research.
Collapse
Affiliation(s)
- Bruce E. Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado Boulder
- BioFrontiers Institute, University of Colorado Boulder
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder
- BioFrontiers Institute, University of Colorado Boulder
- Materials Science and Engineering Program, University of Colorado Boulder
| | | |
Collapse
|
3
|
Pearson JJ, Mao J, Temenoff JS. Effects of Release of TSG-6 from Heparin Hydrogels on Supraspinatus Muscle Regeneration. Tissue Eng Part A 2025; 31:195-207. [PMID: 39556321 DOI: 10.1089/ten.tea.2024.0241] [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: 11/19/2024] Open
Abstract
Muscle degeneration after rotator cuff tendon tear is a significant clinical problem. In these experiments, we developed a poly(ethylene glycol)-based injectable granular hydrogel containing two heparin derivatives (fully sulfated [Hep] and fully desulfated [Hep-]) as well as a matrix metalloproteinase-sensitive peptide to promote sustained release of tumor necrosis factor-stimulated gene 6 (TSG-6) over 14+ days in vivo in a rat model of rotator cuff muscle injury. The hydrogel formulations demonstrated similar release profiles in vivo, thus facilitating comparisons between delivery from heparin derivatives on the level of tissue repair in two different areas of muscle (near the myotendious junction [MTJ] and in the muscle belly [MB]) that have been shown previously to have differing responses to rotator cuff tendon injury. We hypothesized that sustained delivery of TSG-6 would enhance the anti-inflammatory response following rotator cuff injury through macrophage polarization and that release from Hep would potentiate this effect throughout the muscle. Inflammatory/immune cells, satellite cells, and fibroadipogenic progenitor cells were analyzed by flow cytometry 3 and 7 days after injury and hydrogel injection, while metrics of muscle healing were examined via immunohistochemistry up to day 14. Results showed controlled delivery of TSG-6 from Hep caused heightened macrophage response (day 7 macrophages, 4.00 ± 1.85% single cells, M2a, 3.27 ± 1.95% single cells) and increased markers of early muscle regeneration (embryonic heavy chain staining) by day 7, particularly in the MTJ region of the muscle. This work provides a novel strategy for localized, controlled delivery of TSG-6 to enhance muscle healing after rotator cuff tear.
Collapse
Affiliation(s)
- Joseph J Pearson
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, Atlanta, Georgia, USA
| | - Jiahui Mao
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, Atlanta, Georgia, USA
| | - Johnna S Temenoff
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech, Emory University, Atlanta, Georgia, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Widener AE, Roberts A, Phelps EA. Granular Hydrogels for Harnessing the Immune Response. Adv Healthc Mater 2024; 13:e2303005. [PMID: 38145369 PMCID: PMC11196388 DOI: 10.1002/adhm.202303005] [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: 09/08/2023] [Revised: 11/13/2023] [Indexed: 12/26/2023]
Abstract
This review aims to understand the current progress in immune-instructive granular hydrogels and identify the key features used as immunomodulatory strategies. Published work is systematically reviewed and relevant information about granular hydrogels used throughout these studies is collected. The base polymer, microgel generation technique, polymer crosslinking chemistry, particle size and shape, annealing strategy, granular hydrogel stiffness, pore size and void space, degradability, biomolecule presentation, and drug release are cataloged for each work. Several granular hydrogel parameters used for immune modulation: porosity, architecture, bioactivity, drug release, cell delivery, and modularity, are identified. The authors found in this review that porosity is the most significant factor influencing the innate immune response to granular hydrogels, while incorporated bioactivity is more significant in influencing adaptive immune responses. Here, the authors' findings and summarized results from each section are presented and suggestions are made for future studies to better understand the benefits of using immune-instructive granular hydrogels.
Collapse
Affiliation(s)
- Adrienne E Widener
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| | - Abilene Roberts
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| |
Collapse
|
5
|
Daly AC. Granular Hydrogels in Biofabrication: Recent Advances and Future Perspectives. Adv Healthc Mater 2024; 13:e2301388. [PMID: 37317658 DOI: 10.1002/adhm.202301388] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Granular hydrogels, which are formed by densely packing microgels, are promising materials for bioprinting due to their extrudability, porosity, and modularity. However, the multidimensional parameter space involved in granular hydrogel design makes material optimization challenging. For example, design inputs such as microgel morphology, packing density, or stiffness can influence multiple rheological properties that govern printability and the behavior of encapsulated cells. This review provides an overview of fabrication methods for granular hydrogels, and then examines how important design inputs can influence material properties associated with printability and cellular responses across multiple scales. Recent applications of granular design principles in bioink engineering are described, including the development of granular support hydrogels for embedded printing. Further, the paper provides an overview of how key physical properties of granular hydrogels can influence cellular responses, highlighting the advantages of granular materials for promoting cell and tissue maturation after the printing process. Finally, potential future directions for advancing the design of granular hydrogels for bioprinting are discussed.
Collapse
Affiliation(s)
- Andrew C Daly
- Biomedical Engineering, University of Galway, Galway, H91 TK33, Ireland
- CÚRAM the Science Foundation Ireland Research Centre for Medical Devices, University of Galway, Galway, H91 TK33, Ireland
| |
Collapse
|
6
|
Asadikorayem M, Surman F, Weber P, Weber D, Zenobi-Wong M. Zwitterionic Granular Hydrogel for Cartilage Tissue Engineering. Adv Healthc Mater 2024; 13:e2301831. [PMID: 37501337 DOI: 10.1002/adhm.202301831] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Zwitterionic hydrogels have high potential for cartilage tissue engineering due to their ultra-hydrophilicity, nonimmunogenicity, and superior antifouling properties. However, their application in this field has been limited so far, due to the lack of injectable zwitterionic hydrogels that allow for encapsulation of cells in a biocompatible manner. Herein, a novel strategy is developed to engineer cartilage employing zwitterionic granular hydrogels that are injectable, self-healing, in situ crosslinkable and allow for direct encapsulation of cells with biocompatibility. The granular hydrogel is produced by mechanical fragmentation of bulk photocrosslinked hydrogels made of zwitterionic carboxybetaine acrylamide (CBAA), or a mixture of CBAA and zwitterionic sulfobetaine methacrylate (SBMA). The produced microgels are enzymatically crosslinkable using horseradish peroxidase, to quickly stabilize the construct, resulting in a microporous hydrogel. Encapsulated human primary chondrocytes are highly viable and able to proliferate, migrate, and produce cartilaginous extracellular matrix (ECM) in the zwitterionic granular hydrogel. It is also shown that by increasing hydrogel porosity and incorporation of SBMA, cell proliferation and ECM secretion are further improved. This strategy is a simple and scalable method, which has high potential for expanding the versatility and application of zwitterionic hydrogels for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, Zürich, 8032, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| |
Collapse
|
7
|
Lin CH, Srioudom JR, Sun W, Xing M, Yan S, Yu L, Yang J. The use of hydrogel microspheres as cell and drug delivery carriers for bone, cartilage, and soft tissue regeneration. BIOMATERIALS TRANSLATIONAL 2024; 5:236-256. [PMID: 39734701 PMCID: PMC11681182 DOI: 10.12336/biomatertransl.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 12/31/2024]
Abstract
Bone, cartilage, and soft tissue regeneration is a complex process involving many cellular activities across various cell types. Autografts remain the "gold standard" for the regeneration of these tissues. However, the use of autografts is associated with many disadvantages, including donor scarcity, the requirement of multiple surgeries, and the risk of infection. The development of tissue engineering techniques opens new avenues for enhanced tissue regeneration. Nowadays, the expectations of tissue engineering scaffolds have gone beyond merely providing physical support for cell attachment. Ideal scaffolds should also provide biological cues to actively boost tissue regeneration. As a new type of injectable biomaterial, hydrogel microspheres have been increasingly recognised as promising therapeutic carriers for the local delivery of cells and drugs to enhance tissue regeneration. Compared to traditional tissue engineering scaffolds and bulk hydrogel, hydrogel microspheres possess distinct advantages, including less invasive delivery, larger surface area, higher transparency for visualisation, and greater flexibility for functionalisation. Herein, we review the materials characteristics of hydrogel microspheres and compare their fabrication approaches, including microfluidics, batch emulsion, electrohydrodynamic spraying, lithography, and mechanical fragmentation. Additionally, based on the different requirements for bone, cartilage, nerve, skin, and muscle tissue regeneration, we summarize the applications of hydrogel microspheres as cell and drug delivery carriers for the regeneration of these tissues. Overall, hydrogel microspheres are regarded as effective therapeutic delivery carriers to enhance tissue regeneration in regenerative medicine. However, significant effort is required before hydrogel microspheres become widely accepted as commercial products for clinical use.
Collapse
Affiliation(s)
- Chung-Hsun Lin
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jesse R. Srioudom
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Wei Sun
- Leicester International Institute, Dalian University of Technology, Panjin, Liaoning Province, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Canada
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Le Yu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Division of Biological and Biomedical Systems, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Jian Yang
- Biomedical Engineering Program, Westlake University, Hangzhou, Zhejiang Province, China
- Research Centre for Industries of the Future, Westlake University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
8
|
Recalde Phillips S, Perez-Ponce KD, Ruben E, Baig T, Poux E, Gregory CA, Alge DL. Impact of Annealing Chemistry on the Properties and Performance of Microporous Annealed Particle Hydrogels. Biomacromolecules 2024; 25:5798-5808. [PMID: 39190621 PMCID: PMC11388458 DOI: 10.1021/acs.biomac.4c00465] [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: 04/05/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
Microporous annealed particle (MAP) hydrogels are a promising class of in situ-forming scaffolds for tissue repair and regeneration. While an expansive toolkit of annealing chemistries has been described, the effects of different annealing chemistries on MAP hydrogel properties and performance have not been studied. In this study, we address this gap through a controlled head-to-head comparison of poly(ethylene glycol) (PEG)-based MAP hydrogels that were annealed using tetrazine-norbornene and thiol-norbornene click chemistry. Characterization of material properties revealed that tetrazine click annealing significantly increases MAP hydrogel shear storage modulus and results in slower in vitro degradation kinetics when microgels with a higher cross-link density are used. However, these effects are muted when the MAP hydrogels are fabricated from microgels with a lower cross-link density. In contrast, in vivo testing in murine critical-sized calvarial defects revealed that these differences in physicochemical properties do not translate to differences in bone volume or calvarial defect healing when growth-factor-loaded MAP hydrogel scaffolds are implanted into mouse calvarial defects. Nonetheless, the impact of tetrazine click annealing could be important in other applications and should be investigated further.
Collapse
Affiliation(s)
- Sarea
Y. Recalde Phillips
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
| | - Kiara D. Perez-Ponce
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
| | - Elizabeth Ruben
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
| | - Talia Baig
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
| | - Emily Poux
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Carl A. Gregory
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
- Department
of Medical Physiology, School of Medicine, Texas A&M University, Bryan, Texas 77807, United States
| | - Daniel L. Alge
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
- Department
of Materials Science and Engineering, Texas
A&M University, College Station, Texas 77843, United States
| |
Collapse
|
9
|
Khairallah T, Khoury LR. Aided Porous Medium Emulsification for Functional Hydrogel Microparticles Synthesis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311841. [PMID: 39091048 DOI: 10.1002/adma.202311841] [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: 11/08/2023] [Revised: 06/05/2024] [Indexed: 08/04/2024]
Abstract
Despite the substantial advancement in developing various hydrogel microparticle (HMP) synthesis methods, emulsification through porous medium to synthesize functional hybrid protein-polymer HMPs has yet to be addressed. Here, the aided porous medium emulsification for hydrogel microparticle synthesis (APME-HMS) system, an innovative approach drawing inspiration from porous medium emulsification is introduced. This method capitalizes on emulsifying immiscible phases within a 3D porous structure for optimal HMP production. Using the APME-HMS system, synthesized responsive bovine serum albumin (BSA) and polyethylene glycol diacrylate (PEGDA) HMPs of various sizes are successfully synthesized. Preserving protein structural integrity and functionality enable the formation of cytochrome c (cyt c) - PEGDA HMPs for hydrogen peroxide (H2O2) detection at various concentrations. The flexibility of the APME-HMS system is demonstrated by its ability to efficiently synthesize HMPs using low volumes (≈50 µL) and concentrations (100 µm) of proteins within minutes while preserving proteins' structural and functional properties. Additionally, the capability of the APME-HMS method to produce a diverse array of HMP types enriches the palette of HMP fabrication techniques, presenting it as a cost-effective, biocompatible, and scalable alternative for various biomedical applications, such as controlled drug delivery, 3D printing bio-inks, biosensing devices, with potential implications even in culinary applications.
Collapse
Affiliation(s)
- Tina Khairallah
- Department of Materials Science and Engineering, Technion Israel Institute of Technology, Haifa, 32000, Israel
| | - Luai R Khoury
- Department of Materials Science and Engineering, Technion Israel Institute of Technology, Haifa, 32000, Israel
| |
Collapse
|
10
|
Pearson JJ, Mao J, Temenoff JS. Effects of Release of TSG-6 from Heparin Hydrogels on Supraspinatus Muscle Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608812. [PMID: 39229126 PMCID: PMC11370378 DOI: 10.1101/2024.08.20.608812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Muscle degeneration after rotator cuff tendon tear is a significant clinical problem. In these experiments, we developed a poly(ethylene glycol)-based injectable granular hydrogel containing two heparin derivatives (fully sulfated (Hep) and fully desulfated (Hep-)) as well as a matrix metalloproteinase-sensitive peptide to promote sustained release of Tumor Necrosis Factor Stimulated Gene 6 (TSG-6) over 14+ days in vivo in a rat model of rotator cuff muscle injury. The hydrogel formulations demonstrated similar release profiles in vivo , thus facilitating comparisons between delivery from heparin derivatives on level of tissue repair in two different areas of muscle (near the myotendious junction (MTJ) and in the muscle belly (MB)) that have been shown previously to have differing responses to rotator cuff tendon injury. We hypothesized that sustained delivery of TSG-6 would enhance the anti-inflammatory response following rotator cuff injury through macrophage polarization, and that release from a fully sulfated heparin derivative (Hep) would potentiate this effect throughout the muscle. Inflammatory/immune cells, satellite cells, and fibroadipogenic progenitor cells, were analyzed by flow cytometery 3 and 7 days after injury and hydrogel injection, while metrics of muscle healing were examined via immunohistochemistry up to Day 14. Results showed controlled delivery of TSG-6 from Hep caused heightened macrophage response (Day 14 macrophages, 4.00 ± 1.85% single cells, M2a, 3.27 ± 1.95% single cells) and increased markers of early muscle regeneration (embryonic heavy chain staining) by Day 7, particularly in the MTJ region of the muscle, compared to release from desulfated heparin hydrogels. This work provides a novel strategy for localized, controlled delivery of TSG-6 to enhance muscle healing after rotator cuff tear. IMPACT STATEMENT Rotator cuff tear is a significant problem that can cause muscle degeneration. In this study, a hydrogel particle system was developed for sustained release of an anti-inflammatory protein, Tumor Necrosis Factor Stimulated Gene 6 (TSG-6), to injured muscle. Release of the protein from a fully sulfated heparin hydrogel-based carrier demonstrated greater changes in amount inflammatory cells and more early regenerative effects than a less-sulfated carrier. Thus, this work provides a novel strategy for localized, controlled delivery of an anti-inflammatory protein to enhance muscle healing after rotator cuff tear.
Collapse
|
11
|
Chang CY, Nguyen H, Frahm E, Kolaczyk K, Lin CC. Triple click chemistry for crosslinking, stiffening, and annealing of gelatin-based microgels. RSC APPLIED POLYMERS 2024; 2:656-669. [PMID: 39035826 PMCID: PMC11255916 DOI: 10.1039/d3lp00249g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/22/2024] [Indexed: 07/23/2024]
Abstract
Microgels are spherical hydrogels with physicochemical properties ideal for many biomedical applications. For example, microgels can be used as individual carriers for suspension cell culture or jammed/annealed into granular hydrogels with micron-scale pores highly permissive to molecular transport and cell proliferation/migration. Conventionally, laborious optimization processes are often needed to create microgels with different moduli, sizes, and compositions. This work presents a new microgel and granular hydrogel preparation workflow using gelatin-norbornene-carbohydrazide (GelNB-CH). As a gelatin-derived macromer, GelNB-CH presents cell adhesive and degradable motifs while being amenable to three orthogonal click chemistries, namely the thiol-norbornene photo-click reaction, hydrazone bonding, and the inverse electron demand Diels-Alder (iEDDA) click reaction. The thiol-norbornene photo-click reaction (with thiol-bearing crosslinkers) and hydrazone bonding (with aldehyde-bearing crosslinkers) were used to crosslink the microgels and to realize on-demand microgel stiffening, respectively. The tetrazine-norbornene iEDDA click reaction (with tetrazine-bearing crosslinkers) was used to anneal microgels into granular hydrogels. In addition to materials development, we demonstrated the value of the triple-click chemistry granular hydrogels via culturing human mesenchymal stem cells and pancreatic cancer cells.
Collapse
Affiliation(s)
- Chun-Yi Chang
- Weldon School of Biomedical Engineering, Purdue University West Lafayette IN 47907 USA
| | - Han Nguyen
- Weldon School of Biomedical Engineering, Purdue University West Lafayette IN 47907 USA
| | - Ellen Frahm
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis Indianapolis IN 46202 USA
| | - Keith Kolaczyk
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis Indianapolis IN 46202 USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University West Lafayette IN 47907 USA
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis Indianapolis IN 46202 USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center Indianapolis IN 46202 USA
- Integrated Nanosystems Development Institute Indianapolis IN 46202 USA
| |
Collapse
|
12
|
Kan X, Zhang S, Kwok E, Chu Y, Chen L, Zeng X. Granular hydrogels with tunable properties prepared from gum Arabic and protein microgels. Int J Biol Macromol 2024; 273:132878. [PMID: 38844277 DOI: 10.1016/j.ijbiomac.2024.132878] [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: 04/03/2024] [Revised: 05/16/2024] [Accepted: 06/01/2024] [Indexed: 06/17/2024]
Abstract
Granular hydrogels have emerged as a new class of materials for 3D printing, tissue engineering, and food applications due to their extrudability, porosity, and modularity. This work introduces a convenient method to prepare granular hydrogel with tunable properties by modulating the interaction between gum Arabic (GA) and whey protein isolate (WPI) microgels. As the concentration of GA increased, the appearance of the hydrogel changed from fluid liquid to moldable solid, and the microstructure changed from a macro-porous structure with thin walls to a dense structure formed by the accumulation of spherical particles. At a GA concentration of 0.5 %, the hydrogels remained fluid. Granular hydrogels containing 1.0 % GA showed mechanical properties similar to those of tofu (compressive strength: 10.8 ± 0.5 kPa, Young's modulus: 16.7 ± 0.4 kPa), while granular hydrogels containing 1.5 % GA showed mechanical properties similar to those of hawthorn sticks and sausages (compressive strength: 300.4 ± 5.8 kPa; Young's modulus: 200.5 ± 3.4 kPa). The hydrogel with 2.0 % GA was similar to hawthorn sticks, with satisfactory bite resistance and elasticity. Such tunability has led to various application potentials in the food industry to meet consumer demand for healthy, nutritious, and diverse textures.
Collapse
Affiliation(s)
- Xuhui Kan
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Alberta, Canada
| | - Sitian Zhang
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Alberta, Canada
| | - Esther Kwok
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Alberta, Canada
| | - Yifu Chu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Alberta, Canada
| | - Lingyun Chen
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Alberta, Canada.
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| |
Collapse
|
13
|
Xuan L, Hou Y, Liang L, Wu J, Fan K, Lian L, Qiu J, Miao Y, Ravanbakhsh H, Xu M, Tang G. Microgels for Cell Delivery in Tissue Engineering and Regenerative Medicine. NANO-MICRO LETTERS 2024; 16:218. [PMID: 38884868 PMCID: PMC11183039 DOI: 10.1007/s40820-024-01421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/26/2024] [Indexed: 06/18/2024]
Abstract
Microgels prepared from natural or synthetic hydrogel materials have aroused extensive attention as multifunctional cells or drug carriers, that are promising for tissue engineering and regenerative medicine. Microgels can also be aggregated into microporous scaffolds, promoting cell infiltration and proliferation for tissue repair. This review gives an overview of recent developments in the fabrication techniques and applications of microgels. A series of conventional and novel strategies including emulsification, microfluidic, lithography, electrospray, centrifugation, gas-shearing, three-dimensional bioprinting, etc. are discussed in depth. The characteristics and applications of microgels and microgel-based scaffolds for cell culture and delivery are elaborated with an emphasis on the advantages of these carriers in cell therapy. Additionally, we expound on the ongoing and foreseeable applications and current limitations of microgels and their aggregate in the field of biomedical engineering. Through stimulating innovative ideas, the present review paves new avenues for expanding the application of microgels in cell delivery techniques.
Collapse
Affiliation(s)
- Leyan Xuan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yingying Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Jialin Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Kai Fan
- School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| | - Liming Lian
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jianhua Qiu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yingling Miao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hossein Ravanbakhsh
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA.
| | - Mingen Xu
- School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China.
| | - Guosheng Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
14
|
Ryoo H, Kimmel H, Rondo E, Underhill GH. Advances in high throughput cell culture technologies for therapeutic screening and biological discovery applications. Bioeng Transl Med 2024; 9:e10627. [PMID: 38818120 PMCID: PMC11135158 DOI: 10.1002/btm2.10627] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 06/01/2024] Open
Abstract
Cellular phenotypes and functional responses are modulated by the signals present in their microenvironment, including extracellular matrix (ECM) proteins, tissue mechanical properties, soluble signals and nutrients, and cell-cell interactions. To better recapitulate and analyze these complex signals within the framework of more physiologically relevant culture models, high throughput culture platforms can be transformative. High throughput methodologies enable scientists to extract increasingly robust and broad datasets from individual experiments, screen large numbers of conditions for potential hits, better qualify and predict responses for preclinical applications, and reduce reliance on animal studies. High throughput cell culture systems require uniformity, assay miniaturization, specific target identification, and process simplification. In this review, we detail the various techniques that researchers have used to face these challenges and explore cellular responses in a high throughput manner. We highlight several common approaches including two-dimensional multiwell microplates, microarrays, and microfluidic cell culture systems as well as unencapsulated and encapsulated three-dimensional high throughput cell culture systems, featuring multiwell microplates, micromolds, microwells, microarrays, granular hydrogels, and cell-encapsulated microgels. We also discuss current applications of these high throughput technologies, namely stem cell sourcing, drug discovery and predictive toxicology, and personalized medicine, along with emerging opportunities and future impact areas.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Hannah Kimmel
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Evi Rondo
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Gregory H. Underhill
- Bioengineering DepartmentUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
15
|
D’Elia A, Jones OL, Canziani G, Sarkar B, Chaiken I, Rodell CB. Injectable Granular Hydrogels Enable Avidity-Controlled Biotherapeutic Delivery. ACS Biomater Sci Eng 2024; 10:1577-1588. [PMID: 38357739 PMCID: PMC10934254 DOI: 10.1021/acsbiomaterials.3c01906] [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: 12/18/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
Protein therapeutics represent a rapidly growing class of pharmaceutical agents that hold great promise for the treatment of various diseases such as cancer and autoimmune dysfunction. Conventional systemic delivery approaches, however, result in off-target drug exposure and a short therapeutic half-life, highlighting the need for more localized and controlled delivery. We have developed an affinity-based protein delivery system that uses guest-host complexation between β-cyclodextrin (CD, host) and adamantane (Ad, guest) to enable sustained localized biomolecule presentation. Hydrogels were formed by the copolymerization of methacrylated CD and methacrylated dextran. Extrusion fragmentation of bulk hydrogels yielded shear-thinning and self-healing granular hydrogels (particle diameter = 32.4 ± 16.4 μm) suitable for minimally invasive delivery and with a high host capacity for the retention of guest-modified proteins. Bovine serum albumin (BSA) was controllably conjugated to Ad via EDC chemistry without affecting the affinity of the Ad moiety for CD (KD = 12.0 ± 1.81 μM; isothermal titration calorimetry). The avidity of Ad-BSA conjugates was directly tunable through the number of guest groups attached, resulting in a fourfold increase in the complex half-life (t1/2 = 5.07 ± 1.23 h, surface plasmon resonance) that enabled a fivefold reduction in protein release at 28 days. Furthermore, we demonstrated that the conjugation of Ad to immunomodulatory cytokines (IL-4, IL-10, and IFNγ) did not detrimentally affect cytokine bioactivity and enabled their sustained release. Our strategy of avidity-controlled delivery of protein-based therapeutics is a promising approach for the sustained local presentation of protein therapeutics and can be applied to numerous biomedical applications.
Collapse
Affiliation(s)
- Arielle
M. D’Elia
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Olivia L. Jones
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Gabriela Canziani
- Department
of Biochemistry and Molecular Biology, Drexel
University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Biplab Sarkar
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Irwin Chaiken
- Department
of Biochemistry and Molecular Biology, Drexel
University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Christopher B. Rodell
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
16
|
An C, Zhang S, Xu J, Zhang Y, Dou Z, Shao F, Long C, yang J, Wang H, Liu J. The microparticulate inks for bioprinting applications. Mater Today Bio 2024; 24:100930. [PMID: 38293631 PMCID: PMC10825055 DOI: 10.1016/j.mtbio.2023.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/05/2023] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
Three-dimensional (3D) bioprinting has emerged as a groundbreaking technology for fabricating intricate and functional tissue constructs. Central to this technology are the bioinks, which provide structural support and mimic the extracellular environment, which is crucial for cellular executive function. This review summarizes the latest developments in microparticulate inks for 3D bioprinting and presents their inherent challenges. We categorize micro-particulate materials, including polymeric microparticles, tissue-derived microparticles, and bioactive inorganic microparticles, and introduce the microparticle ink formulations, including granular microparticles inks consisting of densely packed microparticles and composite microparticle inks comprising microparticles and interstitial matrix. The formulations of these microparticle inks are also delved into highlighting their capabilities as modular entities in 3D bioprinting. Finally, existing challenges and prospective research trajectories for advancing the design of microparticle inks for bioprinting are discussed.
Collapse
Affiliation(s)
- Chuanfeng An
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Shiying Zhang
- School of Dentistry, Shenzhen University, Shenzhen, 518060, China
| | - Jiqing Xu
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Yujie Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Zhenzhen Dou
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Fei Shao
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Canling Long
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Jianhua yang
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Huanan Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116023, China
| | - Jia Liu
- Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| |
Collapse
|
17
|
Morley CD, Ding EA, Carvalho EM, Kumar S. A Balance between Inter- and Intra-Microgel Mechanics Governs Stem Cell Viability in Injectable Dynamic Granular Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304212. [PMID: 37653580 PMCID: PMC10841739 DOI: 10.1002/adma.202304212] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Injectable hydrogels are increasingly explored for the delivery of cells to tissue. These materials exhibit both liquid-like properties, protecting cells from mechanical stress during injection, and solid-like properties, providing a stable 3D engraftment niche. Many strategies for modulating injectable hydrogels tune liquid- and solid-like material properties simultaneously, such that formulation changes designed to improve injectability can reduce stability at the delivery site. The ability to independently tune liquid- and solid-like properties would greatly facilitate formulation development. Here, such a strategy is presented in which cells are ensconced in the pores between microscopic granular hyaluronic acid (HA) hydrogels (microgels), where elasticity is tuned with static covalent intra-microgel crosslinks and flowability with mechanosensitive adamantane-cyclodextrin (AC) inter-microgel crosslinks. Using the same AC-free microgels as a 3D printing support bath, the location of each cell is preserved as it exits the needle, allowing identification of the mechanism driving mechanical trauma-induced cell death. The microgel AC concentration is varied to find the threshold from microgel yielding- to AC interaction-dominated injectability, and this threshold is exploited to fabricate a microgel with better injection-protecting performance. This delivery strategy, and the balance between intra- and inter-microgel properties it reveals, may facilitate the development of new cell injection formulations.
Collapse
Affiliation(s)
- Cameron D Morley
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
18
|
Lee HP, Davis R, Wang TC, Deo KA, Cai KX, Alge DL, Lele TP, Gaharwar AK. Dynamically Cross-Linked Granular Hydrogels for 3D Printing and Therapeutic Delivery. ACS APPLIED BIO MATERIALS 2023; 6:3683-3695. [PMID: 37584641 PMCID: PMC10863386 DOI: 10.1021/acsabm.3c00337] [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: 05/08/2023] [Accepted: 07/13/2023] [Indexed: 08/17/2023]
Abstract
Granular hydrogels have recently emerged as promising biomaterials for tissue engineering and 3D-printing applications, addressing the limitations of bulk hydrogels while exhibiting desirable properties such as injectability and high porosity. However, their structural stability can be improved with post-injection interparticle cross-linking. In this study, we developed granular hydrogels with interparticle cross-linking through reversible and dynamic covalent bonds. We fragmented photo-cross-linked bulk hydrogels to produce aldehyde or hydrazide-functionalized microgels using chondroitin sulfate. Mixing these microgels facilitated interparticle cross-linking through reversible hydrazone bonds, providing shear-thinning and self-healing properties for injectability and 3D printing. The resulting granular hydrogels displayed high mechanical stability without the need for secondary cross-linking. Furthermore, the porosity and sustained release of growth factors from these hydrogels synergistically enhanced cell recruitment. Our study highlights the potential of reversible interparticle cross-linking for designing injectable and 3D printable therapeutic delivery scaffolds using granular hydrogels. Overall, our study highlights the potential of reversible interparticle cross-linking to improve the structural stability of granular hydrogels, making them an effective biomaterial for use in tissue engineering and 3D-printing applications.
Collapse
Affiliation(s)
- Hung-Pang Lee
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
| | - Ryan Davis
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
| | - Ting-Ching Wang
- Chemical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
| | - Kaivalya A. Deo
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
| | - Kathy Xiao Cai
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
| | - Daniel L. Alge
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
- Material
Science and Engineering, College of Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Tanmay P. Lele
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
- Chemical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
| | - Akhilesh K. Gaharwar
- Biomedical
Engineering, College of Engineering, Texas
A&M University, College
Station, Texas 77843, United States
- Material
Science and Engineering, College of Engineering, Texas A&M University, College Station, Texas 77843, United States
- Interdisciplinary
Graduate Program in Genetics & Genomics, Texas A&M University, College Station, Texas 77843, United States
- Center
for Remote Health Technologies and Systems, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
19
|
Widener AE, Roberts A, Phelps EA. Single versus dual microgel species for forming guest-host microporous annealed particle PEG-MAL hydrogel. J Biomed Mater Res A 2023; 111:1379-1389. [PMID: 37010360 PMCID: PMC10909382 DOI: 10.1002/jbm.a.37540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/03/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023]
Abstract
Inter-particle secondary crosslinks allow microporous annealed particle (MAP) hydrogels to be formed. Methods to introduce secondary crosslinking networks in MAP hydrogels include particle jamming, annealing with covalent bonds, and reversible noncovalent interactions. Here, we investigate the effect of two different approaches to secondary crosslinking of polyethylene glycol (PEG) microgels via reversible guest-host interactions. We generated a dual-particle MAP-PEG hydrogel using two species of PEG microgels, one functionalized with the guest molecule, adamantane, and the other with the host molecule, β-cyclodextrin (Inter-MAP-PEG). In a different approach, a mono-particle MAP-PEG hydrogel was generated using one species of microgel functionalized with both guest and host molecules (Intra-MAP-PEG). The Intra-MAP-PEG formed a homogenous distribution due to the single type of microgels used. We then compared the mechanical properties of these two types of MAP-PEG hydrogels and found that Intra-MAP-PEG resulted in significantly softer gels with lower yield stress. We investigated the effect of intra-particle guest-host interactions through titrated weight percentage and the concentration of functional groups added to the hydrogel. We found that there was an ideal concentration of guest-host molecules that enables intra- and inter-particle guest-host interactions with sufficient covalent crosslinking. Based on these studies, Intra-MAP-PEG provides a homogeneous guest-host hydrogel that is shear-thinning with reversible secondary crosslinking.
Collapse
Affiliation(s)
- Adrienne E. Widener
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Abilene Roberts
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|