1
|
Guan Q, Hou S, Wang K, Li L, Cheng Y, Zheng M, Liu C, Zhao X, Zhou J, Li P, Niu X, Wang L, Fan Y. Micropore structure engineering of injectable granular hydrogels via controlled liquid-liquid phase separation facilitates regenerative wound healing in mice and pigs. Biomaterials 2025; 318:123192. [PMID: 39965423 DOI: 10.1016/j.biomaterials.2025.123192] [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: 10/24/2024] [Revised: 01/22/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
Biomaterials can play a crucial role in facilitating tissue regeneration, but their application is often limited by that they induce scarring rather than complete tissue restoration. Hydrogels with microporous architectures, engineered via 3D printing techniques or particle packing (granular hydrogels), have shown promise in providing a conducive microenvironment for cellular infiltration and favorable immune response. Nonetheless, there is a notably lacking in studies that demonstrate scarless regeneration solely through pore structure engineering. In this study, we demonstrate that optimizing micropore structure of injectable granular hydrogels via controlled liquid-liquid phase separation facilitates scarless wound healing. The building block particles are fabricated by precisely controlling the separation kinetics of two immiscible aqueous phases (gelling and porogenic) and timely arresting phase separation, to generate bicontinuous, hollow or closed porous structure. Employing a murine model, we reveal that the optimized pore structure significantly facilitates mature vascular network boosts pro-regenerative macrophage polarization (M2/M1) and CD4+/Foxp3+ regulatory T cells, culminating in scarless skin regeneration enriched with hair follicles. Moreover, our hydrogels outperform the clinical gold-standard collagen/proteoglycan scaffolds in a porcine model, showcasing superior cell infiltration, epidermal integration, and dermal regeneration. Micropore structure engineering of biomaterials presents a promising and biologics free pathway for tissue regeneration.
Collapse
Affiliation(s)
- Qifeng Guan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115, Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Sen Hou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China.
| | - Kai Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Linhao Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yating Cheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Mingxia Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Chen Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xinbin Zhao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Jin Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ping Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xufeng Niu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Lizhen Wang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115, Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115, Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
2
|
Pradal P, Hardivillé P, Kim JB, Nam SK, Kim SH, Amstad E. 3D Printing of Stiff Photonic Microparticles into Load-Bearing Structures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2501172. [PMID: 40159833 DOI: 10.1002/smll.202501172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/14/2025] [Indexed: 04/02/2025]
Abstract
Structural colors are bright and possess a remarkable resistance to light exposure, humidity, and temperature such that they constitute an environmentally friendly alternative to chemical pigments. Unfortunately, upscaling the production of photonic structures obtained via conventional colloidal self-assembly is challenging because defects often occur during the assembly of larger structures. Moreover, the processing of materials exhibiting structural colors into intricate 3D structures remains challenging. To address these limitations, rigid photonic microparticles are formulated into an ink that can be 3D printed through direct ink writing (DIW) at room temperature to form intricate macroscopic structures possessing locally varying mechanical and optical properties. This is achieved by adding small amounts of soft microgels to the rigid photonic particles. To rigidify the granular structure, a percolating hydrogel network is formed that covalently connects the microgels. The mechanical properties of the resulting photonic granular materials can be adjusted with the composition and volume fraction of the microgels. The potential of this approach is demonstrated by 3D printing a centimeter-sized photonic butterfly and a temperature-responsive photonic material.
Collapse
Affiliation(s)
- Pauline Pradal
- Soft Materials Laboratory, Institute of Materials in École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Paul Hardivillé
- Soft Materials Laboratory, Institute of Materials in École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Jong Bin Kim
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, 19104, USA
| | - Seong Kyeong Nam
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Shin-Hyun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Esther Amstad
- Soft Materials Laboratory, Institute of Materials in École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| |
Collapse
|
3
|
Ding A, Tang F, Alsberg E. 4D Printing: A Comprehensive Review of Technologies, Materials, Stimuli, Design, and Emerging Applications. Chem Rev 2025; 125:3663-3771. [PMID: 40106790 DOI: 10.1021/acs.chemrev.4c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
4D printing is a groundbreaking technology that seamlessly integrates additive manufacturing with smart materials, enabling the creation of multiscale objects capable of changing shapes and/or functions in a controlled and programmed manner in response to applied energy inputs. Printing technologies, mathematical modeling, responsive materials, stimuli, and structural design constitute the blueprint of 4D printing, all of which have seen rapid advancement in the past decade. These advancements have opened up numerous possibilities for dynamic and adaptive structures, finding potential use in healthcare, textiles, construction, aerospace, robotics, photonics, and electronics. However, current 4D printing primarily focuses on proof-of-concept demonstrations. Further development is necessary to expand the range of accessible materials and address fabrication complexities for widespread adoption. In this paper, we aim to deliver a comprehensive review of the state-of-the-art in 4D printing, probing into shape programming, exploring key aspects of resulting constructs including printing technologies, materials, structural design, morphing mechanisms, and stimuli-responsiveness, and elaborating on prominent applications across various fields. Finally, we discuss perspectives on limitations, challenges, and future developments in the realm of 4D printing. While the potential of this technology is undoubtedly vast, continued research and innovation are essential to unlocking its full capabilities and maximizing its real-world impact.
Collapse
Affiliation(s)
- Aixiang Ding
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Fang Tang
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Eben Alsberg
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois 60612, United States
- Departments of Mechanical & Industrial Engineering, Orthopaedic Surgery, and Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, United States
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, Illinois 60612, United States
| |
Collapse
|
4
|
Ahmed MS, Yun S, Kim HY, Ko S, Islam M, Nam KW. Hydrogels and Microgels: Driving Revolutionary Innovations in Targeted Drug Delivery, Strengthening Infection Management, and Advancing Tissue Repair and Regeneration. Gels 2025; 11:179. [PMID: 40136884 PMCID: PMC11942270 DOI: 10.3390/gels11030179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Hydrogels and microgels are emerging as pivotal platforms in biomedicine, with significant potential in targeted drug delivery, enhanced infection management, and tissue repair and regeneration. These gels, characterized by their high water content, unique structures, and adaptable mechanical properties, interact seamlessly with biological systems, making them invaluable for controlled and targeted drug release. In the realm of infection management, hydrogels and microgels can incorporate antimicrobial agents, offering robust defenses against bacterial infections. This capability is increasingly important in the fight against antibiotic resistance, providing innovative solutions for infection prevention in wound dressings, surgical implants, and medical devices. Additionally, the biocompatibility and customizable mechanical properties of these gels make them ideal scaffolds for tissue engineering, supporting the growth and repair of damaged tissues. Despite their promising applications, challenges such as ensuring long-term stability, enhancing therapeutic agent loading capacities, and scaling production must be addressed for widespread adoption. This review explores the current advancements, opportunities, and limitations of hydrogels and microgels, highlighting research and technological directions poised to revolutionize treatment strategies through personalized and regenerative approaches.
Collapse
Affiliation(s)
- Md. Shahriar Ahmed
- Department of Energy & Materials Engineering, Dongguk University, Seoul 04620, Republic of Korea; (M.S.A.)
| | - Sua Yun
- Department of Advanced Battery Convergence Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Hae-Yong Kim
- Department of Advanced Battery Convergence Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sunho Ko
- Department of Advanced Battery Convergence Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Mobinul Islam
- Department of Energy & Materials Engineering, Dongguk University, Seoul 04620, Republic of Korea; (M.S.A.)
| | - Kyung-Wan Nam
- Department of Energy & Materials Engineering, Dongguk University, Seoul 04620, Republic of Korea; (M.S.A.)
| |
Collapse
|
5
|
Thompson GB, Lee J, Kamani KM, Flores-Velasco N, Rogers SA, Harley BAC. Granular hydrogels as brittle yield stress fluids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.639638. [PMID: 40060491 PMCID: PMC11888328 DOI: 10.1101/2025.02.22.639638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
While granular hydrogels are increasingly used in biomedical applications, methods to capture their rheological behavior generally consider shear-thinning and self-healing properties or produce ensemble metrics such as the dynamic moduli. Analytical approaches paired with common oscillatory shear tests can describe not only solid-like and fluid-like behavior of granular hydrogels but also transient characteristics inherent in yielding and unyielding processes. Combining oscillatory shear testing with consideration of Brittility (Bt) via the Kamani-Donley-Rogers (KDR) model, we show granular hydrogels behave as brittle yield stress fluids with complex transient rheology. We quantify steady and transient rheology as a function of microgel (composition; diameter) and granular (packing; droplet heterogeneity) assembly properties for mixtures of polyethylene glycol and gelatin microgels. The KDR model with Bt captures granular hydrogel behavior for a wide range of design parameters, reducing the complex transient rheology to a determination of model parameters. We describe the impact of composition on rheological behavior and model parameters in monolithic and mixed granular hydrogels. The model robustly captures self-healing behavior and reveals granular relaxation time depends on strain amplitude. This quantitative framework is an important step toward rational design of granular hydrogels for applications ranging from injection and in situ stabilization to 3D bioprinting.
Collapse
Affiliation(s)
- G B Thompson
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - J Lee
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - K M Kamani
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - N Flores-Velasco
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - S A Rogers
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - B A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
6
|
Bailey SJ, Eckman N, Brunel ES, Jons CK, Sen S, Appel EA. A thiol-ene click-based strategy to customize injectable polymer-nanoparticle hydrogel properties for therapeutic delivery. Biomater Sci 2025; 13:1323-1334. [PMID: 39898598 PMCID: PMC11789556 DOI: 10.1039/d4bm01315h] [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: 10/01/2024] [Accepted: 12/29/2024] [Indexed: 02/04/2025]
Abstract
Polymer-nanoparticle (PNP) hydrogels are a promising injectable biomaterial platform that has been used for a wide range of biomedical applications including adhesion prevention, adoptive cell delivery, and controlled drug release. By tuning the chemical, mechanical, and erosion properties of injected hydrogel depots, additional control over cell compatibility and pharmaceutical release kinetics may be realized. Here, we employ thiol-ene click chemistry to prepare a library of modified hydroxypropylmethylcellulose (HPMC) derivatives for subsequent use in PNP hydrogel applications. When combined with poly(ethylene glycol)-b-poly(lactic acid) nanoparticles, we demonstrate that systematically altering the hydrophobic, steric, or pi stacking character of HPMC modifications can readily tailor the mechanical properties of PNP hydrogels. Additionally, we highlight the compatibility of the synthetic platform for the incorporation of cysteine-bearing peptides to access PNP hydrogels with improved bioactivity. Finally, through leveraging the tunable physical properties afforded by this method, we show hydrogel retention time in vivo can be dramatically altered without sacrificing mesh size or cargo diffusion rates. This work offers a route to optimize PNP hydrogels for a variety of translational applications and holds promise in the highly tunable delivery of pharmaceuticals and adoptive cells.
Collapse
Affiliation(s)
- Sophia J Bailey
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Noah Eckman
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Elisa S Brunel
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Carolyn K Jons
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Samya Sen
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Eric A Appel
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA, USA
- Woods Institute for the Environment, Stanford University, Stanford, CA, USA
- Department of Pediatrics - Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
7
|
Zhang L, Su L, Wu L, Zhou W, Xie J, Fan Y, Zhou X, Zhou C, Cui Y, Sun J. Versatile hydrogels prepared by microfluidics technology for bone tissue engineering applications. J Mater Chem B 2025; 13:2611-2639. [PMID: 39876639 DOI: 10.1039/d4tb02314e] [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: 01/30/2025]
Abstract
Bone defects are a prevalent issue resulting from various factors, such as trauma, degenerative diseases, congenital disabilities, and the surgical removal of tumors. Current methods for bone regeneration have limitations. In this context, the fusion of tissue engineering and microfluidics has emerged as a promising strategy in the field of bone regeneration. This study describes the classification of microfluidic devices based on the nature of flow and channel type, as well as the materials and techniques required. An overview of microfluidic methods used to prepare hydrogels and the advantages of using these hydrogels in bone tissue engineering (BTE) combining several basic elements of BTE to highlight its advantages is provided. Furthermore, this work emphasizes the benefits of using hydrogels prepared via microfluidics over conventional hydrogels in BTE because of their controlled release of cargo, they can be used for in situ injection, simplify the steps of single-cell encapsulation and have the advantages of high-throughput and precise preparation. Additionally, organ-on-a-chip models fabricated via microfluidics offer a platform for studying cell and tissue behaviors in an authentic and dynamic environment. Moreover, microfluidic devices can be utilized for noninvasive diagnosis and therapy. Finally, this paper summarizes the preclinical and clinical applications of hydrogels prepared via microfluidics for bone regeneration by focusing on their current developmental status, limitations associated with their application, and future challenges, which underscore their potential impacts on advancing regenerative medicine practices.
Collapse
Affiliation(s)
- Luyue Zhang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Liqian Su
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Lina Wu
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Weikai Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jing Xie
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Yi Fan
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Changchun Zhou
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yujia Cui
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jianxun Sun
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Wang J, Zhang Q, Chen L. Microporous annealed particle hydrogels in cell culture, tissue regeneration, and emerging application in cancer immunotherapy. Am J Cancer Res 2025; 15:665-683. [PMID: 40084361 PMCID: PMC11897623 DOI: 10.62347/wrgw4430] [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: 11/07/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Microporous annealed particle (MAP) hydrogels consist of densely crosslinked and annealed hydrogel particles. Compared to common hydrogels, the inherent porosity within and among these hydrogel particles offers interconnected channels for substance exchange in addition to sufficient growth space for cells, thereby forming a three-dimensional culture system that highly mimics the in vivo microenvironment. Such characteristics enable MAP hydrogels to adapt to various requirements of biomedical applications, along with their excellent injectability and mechanical properties. This review initially provides a comprehensive summary of the fabrication methods and material types of MAP hydrogels, alongside an assessment of their mechanical properties and porosity. In vitro studies are evaluated based on the impact of MAP hydrogels on cellular behaviors, focusing on cell proliferation, differentiation, migration, activity, and phenotype. In vivo research highlights the promising applications of MAP hydrogels in tissue regeneration, as well as their innovative use in cancer immunotherapy. Current challenges and future research directions are outlined, underscoring the potential of MAP hydrogels to significantly improve clinical outcomes in cancer treatment and regenerative medicine.
Collapse
Affiliation(s)
- Junjie Wang
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and TechnologyShanghai 200093, China
| | - Qin Zhang
- Medical Engineering Department of Northern Jiangsu People’s HospitalYangzhou 225009, Jiangsu, China
| | - Liwen Chen
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and TechnologyShanghai 200093, China
| |
Collapse
|
9
|
Stornello DE, Kim J, Chen Z, Heaton K, Qazi TH. Controlling Microparticle Aspect Ratio via Photolithography for Injectable Granular Hydrogel Formation and Cell Delivery. ACS Biomater Sci Eng 2025; 11:1242-1252. [PMID: 39788546 PMCID: PMC11817678 DOI: 10.1021/acsbiomaterials.4c02102] [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: 11/09/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Granular hydrogels are injectable and inherently porous biomaterials assembled through the packing of microparticles. These particles typically have a symmetric and spherical shape. However, recent studies have shown that asymmetric particles with high aspect ratios, such as fibers and rods, can significantly improve the mechanics, structure, and cell-guidance ability of granular hydrogels. Despite this, it remains unknown how controlled changes in the particle aspect ratio influence the injectability, porosity, and cell-instructive capabilities of granular hydrogels. Part of the challenge lies in obtaining microparticles with precisely tailored dimensions using fabrication methods such as flow-focusing microfluidics or extrusion fragmentation. In this work, we leveraged facile photolithography and photocurable hyaluronic acid to fabricate rod-shaped microparticles with widths and heights of 130 μm and lengths that varied from 260 to 1300 μm to obtain aspect ratios (ARs) of 2, 4, 6, 8, and 10. All AR microparticles formed porous and injectable granular hydrogels after centrifugation jamming. Interestingly, the longest microparticles neither clogged the needle nor fractured after extrusion from a syringe. This was attributed to a relatively low elastic modulus that permitted microparticle pliability and reversible deformation under shear. Cells (NIH/3T3 fibroblasts) mixed with the jammed microparticles and injected into molds remained viable, adhered to the particles' surface, and showed a significant and rapid rate of proliferation over a period of 7 days compared to bulk hydrogels. The proliferation rate and morphology of the cells were significantly influenced by the particle AR, with higher cell numbers observed with intermediate ARs, likely attributable to the surface area available for cell adhesion. These findings showcase the utility of injectable granular hydrogels made with high-aspect-ratio microparticles for biomedical applications.
Collapse
Affiliation(s)
- Dean E. Stornello
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette 47907-2050, Indiana, United States
| | - Jun Kim
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette 47907-2050, Indiana, United States
| | - Zhiyuan Chen
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette 47907-2050, Indiana, United States
- Department
of Agricultural and Biological Engineering, Purdue University, West Lafayette 47907-2050, Indiana, United States
| | - Kyle Heaton
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette 47907-2050, Indiana, United States
| | - Taimoor H. Qazi
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette 47907-2050, Indiana, United States
| |
Collapse
|
10
|
Pal V, Gupta D, Liu S, Namli I, Rizvi SHA, Yilmaz YO, Haugh L, Gerhard EM, Ozbolat IT. Interparticle Crosslinked Ion-responsive Microgels for 3D and 4D (Bio)printing Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635095. [PMID: 39975099 PMCID: PMC11838323 DOI: 10.1101/2025.01.28.635095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Microgels offer unique advantages over bulk hydrogels due to their improved diffusion limits for oxygen and nutrients. Particularly, stimuli-responsive microgels with inherently bioactive and self-supporting properties emerge as highly promising biomaterials. This study unveils the development of interparticle-crosslinked, self-supporting, ion-responsive microgels tailored for 3D and 4D (bio)printing applications. A novel strategy was proposed to develop microgels that enabled interparticle crosslinking, eliminating the need for filler hydrogels and preserving essential microscale void spaces to support cell migration and vascularization. Additionally, these microgels possessed unique, ion-responsive shrinking behavior primarily by the Hofmeister effect, reversible upon the removal of the stimulus. Two types of microgels, spherical (µS) and random-shaped (µR), were fabricated, with µR exhibiting superior mechanical properties and higher packing density. Fabricated microgel-based constructs supported angiogenesis with tunable vessel size based on interstitial void spaces while demonstrating excellent shear-thinning and self-healing properties and high print fidelity. Various bioprinting techniques were employed and validated using these microgels, including extrusion-based, embedded, intraembedded, and aspiration-assisted bioprinting, facilitating the biofabrication of scalable constructs. Multi-material 4D printing was achieved by combining ion-responsive microgels with non-responsive microgels, enabling programmable shape transformations upon exposure to ionic solutions. Utilizing 4D printing, complex, dynamic structures were generated such as coiling filaments, grippers, and folding sheets, providing a foundation for the development of advanced tissue models and devices for regenerative medicine and soft robotics, respectively.
Collapse
|
11
|
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
|
12
|
Asadikorayem M, Weber P, Surman F, Puiggalí‐Jou A, Zenobi‐Wong M. Foreign Body Immune Response to Zwitterionic and Hyaluronic Acid Granular Hydrogels Made with Mechanical Fragmentation. Adv Healthc Mater 2025; 14:e2402890. [PMID: 39498680 PMCID: PMC11730820 DOI: 10.1002/adhm.202402890] [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: 08/03/2024] [Revised: 10/19/2024] [Indexed: 11/07/2024]
Abstract
Granular hydrogels have recently attracted the attention for diverse tissue engineering applications due to their versatility and modularity. Despite previous studies showing enhanced viability and metabolism of cells encapsulated in these hydrogels, the in vitro immune response and long-term fibrotic response of these scaffolds have not been well characterized. Here, bulk and granular hydrogels are studied based on synthetic zwitterionic (ZI) and natural polysaccharide hyaluronic acid (HA) made with mechanical fragmentation. In vitro, immunomodulatory studies show an increased stimulatory effect of HA granular hydrogels compared to bulk, while both bulk and granular ZI hydrogels do not induce an inflammatory response. Subcutaneous implantation in mice shows that both ZI and HA granular hydrogels resulted in less collagen capsule deposition around implants compared to bulk HA hydrogels 10 weeks after implantation. Moreover, the HA granular hydrogels are infiltrated by host cells, including macrophages and mature blood vessels, in a porosity-dependent manner. However, a large number of cells, including multinucleated giant cells as well as blood vessels, surround bulk and granular ZI hydrogels and are not able to infiltrate. Overall, this study provides new insights on the long-term stability and fibrotic response of granular hydrogels, paving the way for future studies and applications.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - Anna Puiggalí‐Jou
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - Marcy Zenobi‐Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| |
Collapse
|
13
|
Guan Z, Katla SK, Dahanayake V, Bae J. 3D Printable Poly( N-isopropylacrylamide) Microgel Suspensions with Temperature-Dependent Rheological Responses. ACS APPLIED POLYMER MATERIALS 2024; 6:14095-14105. [PMID: 39697841 PMCID: PMC11650633 DOI: 10.1021/acsapm.3c03230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/20/2024] [Accepted: 03/08/2024] [Indexed: 12/20/2024]
Abstract
Microgel suspensions have garnered significant interest in fundamental research due to their phase transition between liquid-like to paste-like behaviors stemming from tunable interparticle and particle-solvent interactions. Particularly, stimuli-responsive microgels undergo faster volume changes in response to external stimuli in comparison to their bulk counterparts, while maintaining their structural integrity. Here, concentrated and diluted suspensions of poly(N-isopropylacrylamide) (PNIPAm) microgels are dispersed to different packing fractions in water for the characterizations of temperature-responsive rheological responses. In the intrinsic volume phase transition (VPT), polymer chains collapse, and microgels shrink to smaller sizes. Additionally, the intermicrogel and microgel-solvent interactions vary in VPT, which results in microgel clusters that significantly affect the linear shear moduli of suspensions. The effect of the temperature ramp rate of PNIPAm microgel suspensions on rheological responses is characterized. Moreover, the effect of the mass fraction of microgels on the relative viscosity of dilute microgel suspensions is investigated. These results shed light on understanding the heating and cooling rate-dependent temperature responsiveness of PNIPAm microgel suspensions, establishing pathways to regulate the rheological characteristics in temperature-responsive microgel-based platforms. Therefore, this work envisions technological advancements in different fields such as drug delivery, tissue engineering, and diagnostic tools.
Collapse
Affiliation(s)
- Zhecun Guan
- Department
of NanoEngineering, University of California
San Diego, La Jolla, California 92093, United States
| | | | | | - Jinhye Bae
- Department
of NanoEngineering, University of California
San Diego, La Jolla, California 92093, United States
- Chemical
Engineering Program, University of California
San Diego, La Jolla, California 92093, United States
- Materials
Science and Engineering Program, University
of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
14
|
Seyedsalehi A, Saeedinejad F, Toro S, Alipanah F, Quint J, Schmidt TA, Samandari M, Tamayol A. Benefits of In Situ Foamed and Printed Porous Scaffolds in Wound Healing. Adv Healthc Mater 2024; 13:e2401944. [PMID: 39246293 DOI: 10.1002/adhm.202401944] [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: 05/26/2024] [Revised: 08/19/2024] [Indexed: 09/10/2024]
Abstract
Macroporous hydrogels have shown significant promise in biomedical applications, particularly regenerative medicine, due to their enhanced nutrient and waste permeability, improved cell permissibility, and minimal immunogenicity. However, traditional methods of generating porous hydrogels require secondary post-processing steps or harmful reagents making simultaneous fabrication with bioactive factors and cells impossible. Therefore, a handheld printer is engineered for facile and continuous generation and deposition of hydrogel foams directly within the skin defect to form defect-specific macroporous scaffolds. Within the handheld system, a temperature-controlled microfluidic homogenizer is coupled with miniaturized liquid and air pumps to mix sterile air with gelatin methacryloyl (GelMA) at the desired ratio. An integrated photocrosslinking unit is then utilized to crosslink the printed foam in situ to form scaffolds with controlled porosity. The system is optimized to form reliable and uniform GelMA foams. The resulting foam scaffolds demonstrate mechanical properties with excellent flexibility making them suitable for wound healing applications. The results of in vitro cell culture on the scaffolds demonstrate significantly increased cellular activity compared to the solid hydrogel. The in vivo printed foam scaffolds enhanced the rate and quality of wound healing in mice with full-thickness wound without the use of biological materials.
Collapse
Affiliation(s)
- Amir Seyedsalehi
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Farnoosh Saeedinejad
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Steven Toro
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Fatemeh Alipanah
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Tannin A Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| |
Collapse
|
15
|
Rovers MM, Rogkoti T, Bakker BK, Bakal KJ, van Genderen MH, Salmeron‐Sanchez M, Dankers PY. Using a Supramolecular Monomer Formulation Approach to Engineer Modular, Dynamic Microgels, and Composite Macrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405868. [PMID: 39463044 PMCID: PMC11636168 DOI: 10.1002/adma.202405868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/07/2024] [Indexed: 10/29/2024]
Abstract
Microgels show advantages over bulk hydrogels due to convenient control over microgel size and composition, and the ability to use microgels to modularly construct larger hierarchical scaffold hydrogel materials. Here, supramolecular chemistry is used to formulate supramolecular polymer, dynamic microgels solely held together by non-covalent interactions. Four-fold hydrogen bonding ureido-pyrimidinone (UPy) monomers with different functionalities are applied to precisely tune microgel properties in a modular way, via variations in monomer concentration, bifunctional crosslinker ratio, and the incorporation of supramolecular dyes and peptides. Functionalization with a bioactive supramolecular cell-adhesive peptide induced selectivity of cells toward the bioactive microgels over non-active, non-functionalized versions. Importantly, the supramolecular microgels can also be applied as microscale building blocks into supramolecular bulk macrogels with tunable dynamic behavior: a robust and weak macrogel, where the micro- and macrogels are composed of similar molecular building blocks. In a robust macrogel, microgels act as modular micro-building blocks, introducing multi-compartmentalization, while in a weak macrogel, microgels reinforce and enhance mechanical properties. This work demonstrates the potential to modularly engineer higher-length-scale structures using small molecule supramolecular monomers, wherein microgels serve as versatile and modular micro-building units.
Collapse
Affiliation(s)
- Maritza M. Rovers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Theodora Rogkoti
- Centre for the Cellular MicroenvironmentUniversity of Glasgow, Advanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
| | - Bram K. Bakker
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Kalpit J. Bakal
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Mechanical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Marcel H.P. van Genderen
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Manuel Salmeron‐Sanchez
- Centre for the Cellular MicroenvironmentUniversity of Glasgow, Advanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona08010Spain
| | - Patricia Y.W. Dankers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| |
Collapse
|
16
|
McCormack A, Porcza LM, Leslie NR, Melchels FPW. Gellan gum-based granular gels as suspension media for biofabrication. PLoS One 2024; 19:e0312726. [PMID: 39602414 PMCID: PMC11602023 DOI: 10.1371/journal.pone.0312726] [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: 08/04/2024] [Accepted: 10/13/2024] [Indexed: 11/29/2024] Open
Abstract
Engineering 3D tissue-like constructs for applications such as regenerative medicine remains a major challenge in biomedical research. Recently, self-healing, viscoplastic fluids have been introduced as suspension media to allow lower viscosity, water-rich bioinks to be printed within them for the fabrication of more biomimetic structures. Here, we present gellan gum granular gels produced through the application of shear during gelation, as a candidate suspension medium. We demonstrate that these granular gels exhibit viscoplasticity over a wide range of temperatures, permitting their use for 3D bioprinting of filaments and droplets at low (4°C) as well as physiological temperatures. These granular gels exhibit very low yield stresses (down to 0.4 Pa) which facilitated printing at print speeds up to 60 mm.s-1. Furthermore, we demonstrate the printing of cell-laden droplets maintained over 7 days to show the potential for multiple days of cell culture, as well as the fabrication of hydrogel features within a crosslinkable version of the suspension medium containing granular gellan gum and gelatine-methacryloyl. The combination of ease of preparation, high printing speed, wide temperature tolerance, and crosslinkability makes this gellan gum sheared through cooling-induced gelation an attractive candidate for suspended biofabrication.
Collapse
Affiliation(s)
- Andrew McCormack
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Laura M. Porcza
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Nicholas R. Leslie
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Ferry P. W. Melchels
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
- Future Industries Institute, University of South Australia Mawson Lakes, Adelaide, Australia
| |
Collapse
|
17
|
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] [Grants] [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
|
18
|
Korbanka L, Kim JA, Sim S. Macroscopic Assembly of Materials with Engineered Bacterial Spores via Coiled-Coil Interaction. ACS Synth Biol 2024; 13:3668-3676. [PMID: 39393788 PMCID: PMC11856349 DOI: 10.1021/acssynbio.4c00468] [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] [Indexed: 10/13/2024]
Abstract
Herein, we report macroscopic materials formed by the assembly of engineered bacterial spores. Spores were engineered by using a T7-driven expression system to display a high density of pH-responsive self-associating proteins on their surface. The engineered surface protein on the spore surface enabled pH-dependent binding at the protein level and enabled the assembly of granular materials. Mechanical properties remained largely constant with changing pH, but erosion stability was pH-dependent in a manner consistent with the pH-dependent interaction between the engineered surface proteins. Our finding utilizes synthetic biology for the design of macroscopic materials and illuminates the impact of coiled-coil interaction across various length scales.
Collapse
Affiliation(s)
- Lucas Korbanka
- Department of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Ju-An Kim
- Department of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Seunghyun Sim
- Department of Chemistry, University of California Irvine, Irvine, California 92697, United States
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, United States
- Department of Biomedical Engineering, University of California Irvine, Irvine, California 92697, United States
- Center for Synthetic Biology, University of California, Irvine, Irvine, California 92697, United States
| |
Collapse
|
19
|
Mihajlovic M, Pásztor-Jánoska DK, Cadenas J, Adrados CS, Andersen CY, Kristensen SG, Lind JU. 3D culture of ovarian follicles in granular and nanofibrillar hydrogels. BIOMATERIALS ADVANCES 2024; 164:213987. [PMID: 39128246 DOI: 10.1016/j.bioadv.2024.213987] [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: 02/10/2024] [Revised: 06/21/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
3D culture of ovarian follicles in hydrogel matrices is an important emerging tool for basic scientific studies as well as clinical applications such as fertility preservation. For optimizing and scaling 3D culture of preantral follicles, there is a need for identifying biomaterial matrices that simplifies and improves the current culture procedures. At present, microencapsulation of follicles in alginate beads is the most commonly used approach. However, this technique involves notable manual handling and is best suited for encapsulation of single or several follicles. As a potential alternative, we here explore the suitability of different particle-based hydrogel matrices, where follicles can easily be introduced in tunable 3D environments, in large numbers. Specifically, we study the growth of secondary murine follicles in microgranular alginate and nanofibrillar cellulose matrices, with and without cell-binding cues, and map follicle growth against the viscoelastic properties of the matrices. We cultured follicles within the particle-based hydrogels for 10 days and continuously monitored their size, survival, and tendency to extrude oocytes. Interestingly, we observed that the diameter of the growing follicles increased significantly in the particle-based matrices, as compared to state-of-the-art alginate micro-encapsulation. On the other hand, the follicles displayed an increased tendency for early oocyte extrusion in the granular matrices, leading to a notable reduction in the number of intact follicles. We propose that this may be caused by impaired diffusion of nutrients and oxygen through thicker matrices, attributable to our experimental setup. Still, our findings suggest that viscoelastic, granular hydrogels represent promising matrices for 3D culture of early-stage ovarian follicles. In particular, these materials may easily be implemented in advanced culturing devices such as micro-perfusion systems.
Collapse
Affiliation(s)
- Marko Mihajlovic
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | | | - Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Cristina Subiran Adrados
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Claus Yding Andersen
- The Fertility Clinic, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Johan Ulrik Lind
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
20
|
Shaik R, Brown J, Xu J, Lamichhane R, Wang Y, Hong Y, Zhang G. Cardiac Matrix-Derived Granular Hydrogel Enhances Cell Function in 3D Culture. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58346-58356. [PMID: 39413287 PMCID: PMC11542188 DOI: 10.1021/acsami.4c12871] [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] [Indexed: 10/18/2024]
Abstract
Hydrogels derived from decellularized porcine myocardial matrix have demonstrated significant potential as therapeutic delivery platforms for promoting cardiac repair after injury. Our previous study developed a fibrin-enriched cardiac matrix hydrogel to enhance its angiogenic capacities. However, the bulk hydrogel structure may limit their full potential in cell delivery. Recently, granular hydrogels have emerged as a promising class of biomaterials, offering unique features such as a highly interconnected porous structure that facilitates nutrient diffusion and enhances cell viability. Several techniques have been developed for fabricating various types of granular hydrogels, among which extrusion fragmentation is particularly appealing due to its adaptability to many types of hydrogels, low cost, and high scalability. In this study, we first confirmed the effects of the bulk cardiac matrix hydrogel on the viability of encapsulated human umbilical vein endothelial cells and human mesenchymal stem cells. We then tested the feasibility of producing granular hydrogels from both cardiac matrix and fibrin-enriched cardiac matrix through cellular cross-linking of microgels fabricated by extrusion fragmentation. Afterward, we examined the roles of the produced granular hydrogels in the embedded cells and cell spheroids. Our in vitro data demonstrate that cardiac matrix-derived granular hydrogels support optimal viability of encapsulated cells and promote sprouting of human mesenchymal stem cell spheroids. Additionally, granular hydrogel derived from fibrin-enriched cardiac matrix accelerates angiogenic sprouting of embedded human mesenchymal stem cell spheroids. The results obtained from this study lay an important foundation for the future exploration of using cardiac matrix-derived granular hydrogels for cardiac cell therapy.
Collapse
Affiliation(s)
- Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jacob Brown
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Rabina Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, State College, Pennsylvania 16801, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
21
|
Asadikorayem M, Brunel LG, Weber P, Heilshorn SC, Zenobi-Wong M. Porosity dominates over microgel stiffness for promoting chondrogenesis in zwitterionic granular hydrogels. Biomater Sci 2024; 12:5504-5520. [PMID: 39347711 PMCID: PMC11441418 DOI: 10.1039/d4bm00233d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/15/2024] [Indexed: 10/01/2024]
Abstract
Granular hydrogels comprised of jammed, crosslinked microgels offer great potential as biomaterial scaffolds for cell-based therapies, including for cartilage tissue regeneration. As stiffness and porosity of hydrogels affect the phenotype of encapsulated cells and the extent of tissue regeneration, the design of tunable granular hydrogels to control and optimize these parameters is highly desirable. We hypothesized that chondrogenesis could be modulated using a granular hydrogel platform based on biocompatible, zwitterionic materials with independent intra- and inter-microgel crosslinking mechanisms. Microgels are made with mechanical fragmentation of photocrosslinked zwitterionic carboxybetaine acrylamide (CBAA) and sulfobetaine methacrylate (SBMA) hydrogels, and secondarily crosslinked in the presence of cells using horseradish peroxide (HRP) to produce cell-laden granular hydrogels. We varied the intra-microgel crosslinking density to produce microgels with varied stiffnesses (1-3 kPa) and swelling properties. These microgels, when resuspended at the same weight fraction and secondarily crosslinked, resulted in granular hydrogels with distinct porosities (5-40%) due to differing swelling properties. The greatest extent of chondrogenesis was achieved in scaffolds with the highest microgel stiffness and highest porosity. However, when scaffold porosity was kept constant and just microgel stiffness varied, cell phenotype and chondrogenesis were similar across scaffolds. These results indicate the dominant role of granular scaffold porosity on chondrogenesis, whereas microgel stiffness appears to play a relatively minor role. These observations are in contrast to cells encapsulated within conventional bulk hydrogels, where stiffness has been shown to significantly affect chondrocyte response. In summary, we introduce chemically-defined, zwitterionic biomaterials to fabricate versatile granular hydrogels allowing for tunable scaffold porosity and microgel stiffness to study and influence chondrogenesis.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Patrick Weber
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
22
|
Alsharabasy AM, Aljaabary A, Farràs P, Pandit A. Engineering hemin-loaded hyaluronan needle-like microparticles with photoprotective properties against UV-induced tissue damage. J Mater Chem B 2024; 12:9639-9655. [PMID: 39194146 DOI: 10.1039/d4tb01529k] [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: 08/29/2024]
Abstract
This study aimed to develop hyaluronan (HA)-based hydrogel microparticles (MPs) loaded with hemin to address the limitations of traditional macroscale hydrogels. The objective is to design MPs such that they can modulate their physicochemical properties. Given the widespread use of ultraviolet C (UVC) light in various industries and the need for protective measures against accidental exposure, this study evaluated the potential of hemin-loaded MPs to protect human dermal fibroblasts from oxidative stress and cell death caused by UVC exposure. Multiple MP formulations were developed and analysed for size, surface charge, swelling behaviour, degradation rate, and radical scavenging capabilities, both with and without hemin loading. The most promising formulations were tested against UVC-exposed cells to assess cell viability, intracellular nitric oxide (˙NO) and reactive oxygen species levels, and protein carbonylation. The fabricated particles were in the form of microneedles, and the degree of their crosslinking and the role of hemin in the chemical crosslinking reaction were found to influence the surface charge and hydrodynamic diameter of the MPs. Increased crosslinking resulted in reduced swelling, slower degradation, and decreased hemin release rate. MPs with a higher degree of swelling were capable of releasing hemin into the culture medium, leading to enhanced bilirubin generation in dermal fibroblasts following cellular uptake. Pre-treatment with these MPs protected the cells from UVC-induced cell death, nitrosative stress, and protein carbonylation. These findings highlight the potential of the studied MPs to release hemin and to minimise the harmful effects of UVC on dermal fibroblasts.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY, Ireland.
| | - Amal Aljaabary
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY, Ireland.
| | - Pau Farràs
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY, Ireland.
- School of Biological and Chemical Sciences, Ryan Institute, University of Galway, H91 TK33, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY, Ireland.
| |
Collapse
|
23
|
Tanner GI, Schiltz L, Narra N, Figueiredo ML, Qazi TH. Granular Hydrogels Improve Myogenic Invasion and Repair after Volumetric Muscle Loss. Adv Healthc Mater 2024; 13:e2303576. [PMID: 38329892 DOI: 10.1002/adhm.202303576] [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: 10/17/2023] [Revised: 12/26/2023] [Indexed: 02/10/2024]
Abstract
Skeletal muscle injuries including volumetric muscle loss (VML) lead to excessive tissue scarring and permanent functional disability. Despite its high prevalence, there is currently no effective treatment for VML. Bioengineering interventions such as biomaterials that fill the VML defect to support cell and tissue growth are a promising therapeutic strategy. However, traditional biomaterials developed for this purpose lack the pore features needed to support cell infiltration. The present study investigates for the first time, the impact of granular hydrogels on muscle repair - hypothesizing that their flowability will permit conformable filling of the defect site and their inherent porosity will support the invasion of native myogenic cells, leading to effective muscle repair. Small and large microparticle fragments are prepared from photocurable hyaluronic acid polymer via extrusion fragmentation and facile size sorting. In assembled granular hydrogels, particle size and degree of packing significantly influence pore features, rheological behavior, and injectability. Using a mouse model of VML, it is demonstrated that, in contrast to bulk hydrogels, granular hydrogels support early-stage (satellite cell invasion) and late-stage (myofiber regeneration) muscle repair processes. Together, these results highlight the promising potential of injectable and porous granular hydrogels in supporting endogenous repair after severe muscle injury.
Collapse
Affiliation(s)
- Gabrielle I Tanner
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Leia Schiltz
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Niharika Narra
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Marxa L Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
24
|
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
|
25
|
Brunel LG, Christakopoulos F, Kilian D, Cai B, Hull SM, Myung D, Heilshorn SC. Embedded 3D Bioprinting of Collagen Inks into Microgel Baths to Control Hydrogel Microstructure and Cell Spreading. Adv Healthc Mater 2024; 13:e2303325. [PMID: 38134346 PMCID: PMC11192865 DOI: 10.1002/adhm.202303325] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/14/2023] [Indexed: 12/24/2023]
Abstract
Microextrusion-based 3D bioprinting into support baths has emerged as a promising technique to pattern soft biomaterials into complex, macroscopic structures. It is hypothesized that interactions between inks and support baths, which are often composed of granular microgels, can be modulated to control the microscopic structure within these macroscopic-printed constructs. Using printed collagen bioinks crosslinked either through physical self-assembly or bioorthogonal covalent chemistry, it is demonstrated that microscopic porosity is introduced into collagen inks printed into microgel support baths but not bulk gel support baths. The overall porosity is governed by the ratio between the ink's shear viscosity and the microgel support bath's zero-shear viscosity. By adjusting the flow rate during extrusion, the ink's shear viscosity is modulated, thus controlling the extent of microscopic porosity independent of the ink composition. For covalently crosslinked collagen, printing into support baths comprised of gelatin microgels (15-50 µm) results in large pores (≈40 µm) that allow human corneal mesenchymal stromal cells (MSCs) to readily spread, while control samples of cast collagen or collagen printed in non-granular support baths do not allow cell spreading. Taken together, these data demonstrate a new method to impart controlled microscale porosity into 3D printed hydrogels using granular microgel support baths.
Collapse
Affiliation(s)
- Lucia G. Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Fotis Christakopoulos
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - David Kilian
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Sarah M. Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - David Myung
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Li H, Iyer KS, Bao L, Zhai J, Li JJ. Advances in the Development of Granular Microporous Injectable Hydrogels with Non-spherical Microgels and Their Applications in Tissue Regeneration. Adv Healthc Mater 2024; 13:e2301597. [PMID: 37499268 DOI: 10.1002/adhm.202301597] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/03/2023] [Indexed: 07/29/2023]
Abstract
Granular microporous hydrogels are emerging as effective biomaterial scaffolds for tissue engineering due to their improved characteristics compared to traditional nanoporous hydrogels, which better promote cell viability, cell migration, cellular/tissue infiltration, and tissue regeneration. Recent advances have resulted in the development of granular hydrogels made of non-spherical microgels, which compared to those made of spherical microgels have higher macroporosity, more stable mechanical properties, and better ability to guide the alignment and differentiation of cells in anisotropic tissue. The development of these hydrogels as an emerging research area is attracting increasing interest in regenerative medicine. This review first summarizes the fabrication techniques available for non-spherical microgels with different aspect-ratios. Then, it introduces the development of granular microporous hydrogels made of non-spherical microgels, their physicochemical characteristics, and their applications in tissue regeneration. The limitations and future outlook of research on microporous granular hydrogels are also critically discussed.
Collapse
Affiliation(s)
- Haiyan Li
- Chemical and Environment Engineering Department, School of Engineering, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia
| | - Keerthi Subramanian Iyer
- Chemical and Environment Engineering Department, School of Engineering, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia
| | - Lei Bao
- Chemical and Environment Engineering Department, School of Engineering, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia
| | - Jiali Zhai
- School of Science, STEM College, RMIT University, 124 La Trobe Street, Melbourne, VIC, 3001, Australia
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| |
Collapse
|
29
|
Muir VG, Fainor M, Orozco BS, Hilliard RL, Boyes M, Smith HE, Mauck RL, Schaer TP, Burdick JA, Gullbrand SE. Injectable Radiopaque Hyaluronic Acid Granular Hydrogels for Intervertebral Disc Repair. Adv Healthc Mater 2024; 13:e2303326. [PMID: 38142300 PMCID: PMC11193841 DOI: 10.1002/adhm.202303326] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/10/2023] [Indexed: 12/25/2023]
Abstract
Injectable hydrogels offer minimally-invasive treatment options for degenerative disc disease, a prevalent condition affecting millions annually. Many hydrogels explored for intervertebral disc (IVD) repair suffer from weak mechanical integrity, migration issues, and expulsion. To overcome these limitations, an injectable and radiopaque hyaluronic acid granular hydrogel is developed. The granular structure provides easy injectability and low extrusion forces, while the radiopacity enables direct visualization during injection into the disc and non-invasive monitoring after injection. The radiopaque granular hydrogel is injected into rabbit disc explants to investigate restoration of healthy disc mechanics following needle puncture injury ex vivo and then delivered in a minimally-invasive manner into the intradiscal space in a clinically-relevant in vivo large animal goat model of IVD degeneration initiated through degradation by chondroitinase. The radiopaque granular hydrogel successfully halted loss of disc height due to degeneration. Further, the hydrogel not only enhanced proteoglycan content and reduced collagen content in the nucleus pulposus (NP) region compared to degenerative discs, but also helped to maintain the structural integrity of the disc and promote healthy segregation of the NP and annulus fibrosus regions. Overall, this study demonstrates the great potential of an injectable radiopaque granular hydrogel for treatment of degenerative disc disease.
Collapse
Affiliation(s)
- Victoria G Muir
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew Fainor
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Brianna S Orozco
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Rachel L Hilliard
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Madeline Boyes
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harvey E Smith
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Robert L Mauck
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Thomas P Schaer
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Department of Chemical and Biological Engineering, College of Engineering and Applied Science, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Sarah E Gullbrand
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| |
Collapse
|
30
|
Feliciano AJ, Alaoui Selsouli Y, Habibovic P, Birgani ZNT, Moroni L, Baker MB. Granular polyrotaxane microgels as injectable hydrogels for corneal tissue regeneration. Biomater Sci 2024; 12:4993-5009. [PMID: 39169887 DOI: 10.1039/d4bm00409d] [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: 08/23/2024]
Abstract
Corneal diseases, a leading cause of global vision impairment, present challenges in treatment due to corneal tissue donor scarcity and transplant rejection. Hydrogel biomaterials in the form of corneal implants for tissue regeneration, while promising, have faced obstacles related to cellular and tissue integration. This study develops and investigates the potential of granular polyrotaxane (GPR) hydrogels as a scaffold for corneal keratocyte growth and transparent tissue generation. Employing host-guest driven supramolecular interactions, we developed injectable, cytocompatible hydrogels. By optimizing cyclodextrin (CD) concentrations in thiol-ene crosslinked PEG microgels, we observed improved mechanical properties and thermoresponsiveness while preserving injectability. These microgels, adaptable for precise defect filling, 3D printing or tissue culture facilitate enhanced cellular integration with corneal keratocytes and exhibit tissue-like structures in culture. Our findings demonstrate the promise of GPR hydrogels as a minimally invasive avenue for corneal tissue regeneration. These results have the potential to address transplantation challenges, enhance clinical outcomes, and restore vision.
Collapse
Affiliation(s)
- Antonio J Feliciano
- Department of Complex Tissue Regeneration, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands.
| | - Yousra Alaoui Selsouli
- Department of Instructive Biomaterial Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterial Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands
| | - Zeinab Niloofar Tahmasebi Birgani
- Department of Complex Tissue Regeneration, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands.
- Department of Instructive Biomaterial Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands.
| | - Matthew B Baker
- Department of Complex Tissue Regeneration, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands.
- Department of Instructive Biomaterial Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands
| |
Collapse
|
31
|
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
|
32
|
Arjunan KK, Weng CY, Sheng YJ, Tsao HK. Formation of Self-Healing Granular Eutectogels through Jammed Carbopol Microgels in Supercooled Deep Eutectic Solvent. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:17081-17089. [PMID: 39078642 PMCID: PMC11325637 DOI: 10.1021/acs.langmuir.4c02069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Typically, gel-like materials consist of a polymer network structure in a solvent. In this work, a gel-like material is developed in a deep eutectic solvent (DES) without the presence of a polymer network, achieved simply by adding microgels. The DES is composed of choline chloride and citric acid and remains stably in a supercooled state at room temperature, exhibiting Newtonian fluid behavior with high viscosity. When the microgel (Carbopol) concentration exceeds 2 wt %, the DES undergoes a transition from a liquid to a soft gel state, characterized as a granular eutectogel. The soft gel characteristics of eutectogels exhibit a yield stress, and their storage moduli exceed the loss moduli. The yield stress and storage moduli are observed to increase with increasing microgel concentration. In contrast, the ion conductivity decreases with increasing microgel concentration but eventually levels off. Because the eutectogel can dissolve completely in excess water, it is a physical gel-like material, attributed to the densely packed structure of microgels in the supercooled DES. Due to the absence of networks, the granular eutectogel has the capability to self-heal simply by being pushed together after being cut into two pieces.
Collapse
Affiliation(s)
- Karthi Keyan Arjunan
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan
| | - Chun-Yun Weng
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan
| | - Yu-Jane Sheng
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Heng-Kwong Tsao
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan
| |
Collapse
|
33
|
Zhang Y, Chen J, Sun Y, Wang M, Liu H, Zhang W. Endogenous Tissue Engineering for Chondral and Osteochondral Regeneration: Strategies and Mechanisms. ACS Biomater Sci Eng 2024; 10:4716-4739. [PMID: 39091217 DOI: 10.1021/acsbiomaterials.4c00603] [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: 08/04/2024]
Abstract
Increasing attention has been paid to the development of effective strategies for articular cartilage (AC) and osteochondral (OC) regeneration due to their limited self-reparative capacities and the shortage of timely and appropriate clinical treatments. Traditional cell-dependent tissue engineering faces various challenges such as restricted cell sources, phenotypic alterations, and immune rejection. In contrast, endogenous tissue engineering represents a promising alternative, leveraging acellular biomaterials to guide endogenous cells to the injury site and stimulate their intrinsic regenerative potential. This review provides a comprehensive overview of recent advancements in endogenous tissue engineering strategies for AC and OC regeneration, with a focus on the tissue engineering triad comprising endogenous stem/progenitor cells (ESPCs), scaffolds, and biomolecules. Multiple types of ESPCs present within the AC and OC microenvironment, including bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived mesenchymal stem cells (AD-MSCs), synovial membrane-derived mesenchymal stem cells (SM-MSCs), and AC-derived stem/progenitor cells (CSPCs), exhibit the ability to migrate toward injury sites and demonstrate pro-regenerative properties. The fabrication and characteristics of scaffolds in various formats including hydrogels, porous sponges, electrospun fibers, particles, films, multilayer scaffolds, bioceramics, and bioglass, highlighting their suitability for AC and OC repair, are systemically summarized. Furthermore, the review emphasizes the pivotal role of biomolecules in facilitating ESPCs migration, adhesion, chondrogenesis, osteogenesis, as well as regulating inflammation, aging, and hypertrophy-critical processes for endogenous AC and OC regeneration. Insights into the applications of endogenous tissue engineering strategies for in vivo AC and OC regeneration are provided along with a discussion on future perspectives to enhance regenerative outcomes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
34
|
Mathews HF, Çeper T, Speen T, Bastard C, Bulut S, Pieper MI, Schacher FH, De Laporte L, Pich A. Engineering poly(dehydroalanine)-based gels via droplet-based microfluidics: from bulk to microspheres. SOFT MATTER 2024; 20:6231-6246. [PMID: 39051502 DOI: 10.1039/d4sm00676c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Biomedical applications such as drug delivery, tissue engineering, and functional surface coating rely on switchable adsorption and desorption of specialized guest molecules. Poly(dehydroalanine), a polyzwitterion containing pH-dependent positive and negative charges, shows promise for such reversible loading, especially when integrated into a gel network. Herein, we present the fabrication of poly(dehydroalanine)-derived gels of different size scales and evaluate them with respect to their practical use in biomedicine. Already existing protocols for bulk gelation were remodeled to derive suitable reaction conditions for droplet-based microfluidic synthesis. Depending on the layout of the microfluidic chip, microgels with a size of approximately 30 μm or 200 μm were obtained, whose crosslinking density can be increased by implementing a multi-arm crosslinker. We analyzed the effects of the crosslinker species on composition, permeability, and softness and show that the microgels exhibit advantageous properties inherent to zwitterionic polymer systems, including high hydrophilicity as well as pH- and ionic strength-sensitivity. We demonstrate pH-regulated uptake and release of fluorescent model dyes before testing the adsorption of a small antimicrobial peptide, LL-37. Quantification of the peptide accommodated within the microgels reveals the impact of size and crosslinking density of the microgels. Biocompatibility of the microgels was validated by cell tests.
Collapse
Affiliation(s)
- Hannah F Mathews
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
| | - Tolga Çeper
- Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University Jena, Humboldtstr. 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
- Center for Energy and Environmental Chemistry Jena (CEEC), Friedrich-Schiller-University Jena, Philosophenweg 7a, 07743 Jena, Germany
| | - Tobias Speen
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
| | - Céline Bastard
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
| | - Selin Bulut
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
| | - Maria I Pieper
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
| | - Felix H Schacher
- Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University Jena, Humboldtstr. 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743 Jena, Germany
- Center for Energy and Environmental Chemistry Jena (CEEC), Friedrich-Schiller-University Jena, Philosophenweg 7a, 07743 Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Grüne Aue, 07754 Jena, Germany
| | - Laura De Laporte
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
- Institute of Applied Medical Engineering (AME), Department of Advanced Materials for Biomedicine (AMB), University Hospital RWTH Aachen, Center for Biohybrid Medical Systems (CMBS), Forckenbeckstr. 55, 52074 Aachen, Germany
| | - Andrij Pich
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringer Weg 2, 52074 Aachen, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Brightland Chemelot Campus, Maastricht University, 6167 RD Geleen, The Netherlands
| |
Collapse
|
35
|
Li S, Yang J. Double-crosslinked self-healing hydrogel alleviates osteoarthritis by protecting from wearing and targeting NF-kB signaling. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1879-1891. [PMID: 38860745 DOI: 10.1080/09205063.2024.2360759] [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: 01/16/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024]
Abstract
Osteoarthritis (OA) is a chronic disease that causes pain, morbidity, and disability. The main strategy for OA treatment focuses on inflammation suppression, inhibition of osteoclastogenesis, and protection of articular cartilage. These functions cannot be performed effectively by monotherapy. Therefore, an effective drug delivery system is required, capable of containing and controlling the efflux of various drugs to alleviate osteoclastogenesis, protect cartilage and subchondral bone, and suppress inflammation. In this work, an encapsulation system is constructed using a self-healing chitosan hydrogel and allocated compound drugs. The self-healing gel is composed of branched-functionalized chitosan, created by simultaneously using polycaprolactone polyethylene glycol azide as a block polymer and the host-guest assembly of β-cyclodextrin and adamantane. Inhibitors of the NFkB pathway are loaded into the cavities of β-cyclodextrin and the spring-like structure of the block polymer, which can be rapidly released upon joint friction (due to the reassembly of β-cyclodextrin and adamantane by shear stress and the stretch of the block polymer). In vitro experiments using BMMs and the ATDC5 cell line confirm that the developed hydrogel can simultaneously suppress osteoclastogenesis and induce chondrogenesis. Additionally, a model of knee arthritis in C57 mice was used to confirm that this double-crosslinked encapsulation system can lubricate the knee joint surface and provide adequate protection on demand through shear-responsive drug release.
Collapse
Affiliation(s)
- Shengyun Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, China
| | - Jie Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
36
|
Aizik G, Ostertag-Hill CA, Chakraborty P, Choi W, Pan M, Mankus DV, Lytton-Jean AKR, Kohane DS. Injectable hydrogel based on liposome self-assembly for controlled release of small hydrophilic molecules. Acta Biomater 2024; 183:101-110. [PMID: 38834149 PMCID: PMC11239275 DOI: 10.1016/j.actbio.2024.05.044] [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: 11/28/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Controlled release of low molecular weight hydrophilic drugs, administered locally, allows maintenance of high concentrations at the target site, reduces systemic side effects, and improves patient compliance. Injectable hydrogels are commonly used as a vehicle. However, slow release of low molecular weight hydrophilic drugs is very difficult to achieve, mainly due to a rapid diffusion of the drug out of the drug delivery system. Here we present an injectable and self-healing hydrogel based entirely on the self-assembly of liposomes. Gelation of liposomes, without damaging their structural integrity, was induced by modifying the cholesterol content and surface charge. The small hydrophilic molecule, sodium fluorescein, was loaded either within the extra-liposomal space or encapsulated into the aqueous cores of the liposomes. This encapsulation strategy enabled the achievement of controlled and adjustable release profiles, dependent on the mechanical strength of the gel. The hydrogel had a high mechanical strength, minimal swelling, and slow degradation. The liposome-based hydrogel had prolonged mechanical stability in vivo with benign tissue reaction. This work presents a new class of injectable hydrogel that holds promise as a versatile drug delivery system. STATEMENT OF SIGNIFICANCE: The porous nature of hydrogels poses a challenge for delivering small hydrophilic drug, often resulting in initial burst release and shorten duration of release. This issue is particularly pronounced with physically crosslinked hydrogels, since their matrix can swell and dissipate rapidly, but even in cases where the polymers in the hydrogel are covalently cross-linked, small molecules can be rapidly released through its porous mesh. Here we present an injectable self-healing hydrogel based entirely on the self-assembly of liposomes. Small hydrophilic molecules were entrapped inside the extra-liposomal space or loaded into the aqueous cores of the liposomes, allowing controlled and tunable release profiles.
Collapse
Affiliation(s)
- Gil Aizik
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Claire A Ostertag-Hill
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Priyadarshi Chakraborty
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wonmin Choi
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michelle Pan
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David V Mankus
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Abigail K R Lytton-Jean
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Boston Children's Hospital, Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA; Department of Anesthesiology, Critical Care, and Pain Management, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Rijns L, Baker MB, Dankers PYW. Using Chemistry To Recreate the Complexity of the Extracellular Matrix: Guidelines for Supramolecular Hydrogel-Cell Interactions. J Am Chem Soc 2024; 146:17539-17558. [PMID: 38888174 PMCID: PMC11229007 DOI: 10.1021/jacs.4c02980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Hydrogels have emerged as a promising class of extracellular matrix (ECM)-mimicking materials in regenerative medicine. Here, we briefly describe current state-of-the-art of ECM-mimicking hydrogels, ranging from natural to hybrid to completely synthetic versions, giving the prelude to the importance of supramolecular interactions to make true ECM mimics. The potential of supramolecular interactions to create ECM mimics for cell culture is illustrated through a focus on two different supramolecular hydrogel systems, both developed in our laboratories. We use some recent, significant findings to present important design principles underlying the cell-material interaction. To achieve cell spreading, we propose that slow molecular dynamics (monomer exchange within fibers) is crucial to ensure the robust incorporation of cell adhesion ligands within supramolecular fibers. Slow bulk dynamics (stress-relaxation─fiber rearrangements, τ1/2 ≈ 1000 s) is required to achieve cell spreading in soft gels (<1 kPa), while gel stiffness overrules dynamics in stiffer gels. Importantly, this resonates with the findings of others which specialize in different material types: cell spreading is impaired in case substrate relaxation occurs faster than clutch binding and focal adhesion lifetime. We conclude with discussing considerations and limitations of the supramolecular approach as well as provide a forward thinking perspective to further understand supramolecular hydrogel-cell interactions. Future work may utilize the presented guidelines underlying cell-material interactions to not only arrive at the next generation of ECM-mimicking hydrogels but also advance other fields, such as bioelectronics, opening up new opportunities for innovative applications.
Collapse
Affiliation(s)
- Laura Rijns
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Matthew B. Baker
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology
Inspired Regenerative Medicine, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - Patricia Y. W. Dankers
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Chemical Engineering and Chemistry, Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
38
|
Wang X, Zeng Y, Yuan Z, Chen F, Lo WK, Yuan Y, Li T, Yan X, Wang S. Forced capillary wetting of viscoelastic fluids. J Colloid Interface Sci 2024; 662:555-562. [PMID: 38367573 DOI: 10.1016/j.jcis.2024.02.078] [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: 11/17/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024]
Abstract
HYPOTHESIS Achieving rapid capillary wetting is highly desirable in nature and industries. Previous endeavors have primarily concentrated on passive wetting strategies through surface engineering. However, these approaches are inadequate for high-viscosity fluids due to the significant viscous resistance, especially for non-Newtonian fluids. In contrast, forced wetting emerges as a promising method to address the challenges associated with achieving rapid wetting of non-Newtonian fluids in capillaries. EXPERIMENTS To investigate the forced wetting behavior of viscoelastic fluids in capillaries, we employ Xanthan Gum (XG) aqueous solutions as target fluids with the storage modulus significantly exceeding the loss modulus. We utilize smooth glass capillaries connected to a syringe pump to achieve high moving speeds of up to 1 m/s. FINDINGS Our experiments reveal a significant distinction in the power-law exponent that governs the scaling relationship between the dynamic contact angle and velocity for viscoelastic fluids compared to Newtonian fluids. This exponent is considerably smaller and varies based on the concentration of viscoelastic fluids and the diameter of the capillaries. We suggest that the viscosity dominates the wetting dynamics of viscoelastic fluids, manifested by the contact line morphology-dependent behavior. This insight has significant implications for microfluidics and drug injectability.
Collapse
Affiliation(s)
- Xiong Wang
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China; Centre for Nature-Inspired Engineering, City University of Hong Kong, Hong Kong, China.
| | - Yijun Zeng
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| | - Zhenyue Yuan
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| | - Feipeng Chen
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China
| | - Wai Kin Lo
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China; Centre for Nature-Inspired Engineering, City University of Hong Kong, Hong Kong, China
| | - Yongjiu Yuan
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China; Centre for Nature-Inspired Engineering, City University of Hong Kong, Hong Kong, China
| | - Tong Li
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China; Centre for Nature-Inspired Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiao Yan
- School of Energy and Power Engineering, Chongqing University, Chongqing, China
| | - Steven Wang
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China; Centre for Nature-Inspired Engineering, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
39
|
Ryoo H, Giovanni R, Kimmel H, Jain I, Underhill GH. Combinatorial Microgels for 3D ECM Screening and Heterogeneous Microenvironmental Culture of Primary Human Hepatic Stellate Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303128. [PMID: 38348560 PMCID: PMC11022709 DOI: 10.1002/advs.202303128] [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: 05/22/2023] [Revised: 11/03/2023] [Indexed: 02/15/2024]
Abstract
Nonalcoholic fatty liver disease affects 30% of the United States population and its progression can lead to nonalcoholic steatohepatitis (NASH), and increased risks for cirrhosis and hepatocellular carcinoma. NASH is characterized by a highly heterogeneous liver microenvironment created by the fibrotic activity of hepatic stellate cells (HSCs). While HSCs have been widely studied in 2D, further advancements in physiologically relevant 3D culture platforms for the in vitro modeling of these heterogeneous environments are needed. In this study, the use of stiffness-variable, extracellular matrix (ECM) protein-conjugated polyethylene glycol microgels as 3D cell culture scaffolds to modulate HSC activation is demonstrated. These microgels as a high throughput ECM screening system to identify HSC matrix remodeling and metabolic activities in distinct heterogeneous microenvironmental conditions are further employed. The 6 kPa fibronectin microgels are shown to significantly increase HSC matrix remodeling and metabolic activities in single or multiple-component microenvironments. Overall, heterogeneous microenvironments consisting of multiple distinct ECM microgels promoted a decrease in HSC matrix remodeling and metabolic activities compared to homogeneous microenvironments. The study envisions this ECM screening platform being adapted to a broad number of cell types to aid the identification of ECM microenvironments that best recapitulate the desired phenotype, differentiation, or drug efficacy.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Regina Giovanni
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Hannah Kimmel
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Ishita Jain
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Gregory H. Underhill
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
40
|
Carvalho BG, Nakayama A, Miwa H, Han SW, de la Torre LG, Di Carlo D, Lee J, Kim HJ, Khademhosseini A, de Barros NR. Gelatin methacryloyl granular scaffolds for localized mRNA delivery. AGGREGATE (HOBOKEN, N.J.) 2024; 5:e464. [PMID: 38800607 PMCID: PMC11126212 DOI: 10.1002/agt2.464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
mRNA therapy is the intracellular delivery of messenger RNA (mRNA) to produce desired therapeutic proteins. Developing strategies for local mRNA delivery is still required where direct intra-articular injections are inappropriate for targeting a specific tissue. The mRNA delivery efficiency depends on protecting nucleic acids against nuclease-mediated degradation and safe site-specific intracellular delivery. Herein, we report novel mRNA-releasing matrices based on RGD-moiety-rich gelatin methacryloyl (GelMA) microporous annealed particle (MAP) scaffolds. GelMA concentration in aerogel-based microgels (μgels) produced through a microfluidic process, MAP stiffnesses, and microporosity are crucial parameters for cell adhesion, spreading, and proliferation. After being loaded with mRNA complexes, MAP scaffolds composed of 10 % GelMA μgels display excellent cell viability with increasing cell infiltration, adhesion, proliferation, and gene transfer. The intracellular delivery is achieved by the sustained release of mRNA complexes from MAP scaffolds and cell adhesion on mRNA-releasing scaffolds. These findings highlight that hybrid systems can achieve efficient protein expression by delivering mRNA complexes, making them promising mRNA-releasing biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Bruna Gregatti Carvalho
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Brazil; Terasaki Institute for Biomedical Innovation (TIBI), 90064, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation (TIBI), 90064, USA
| | - Hiromi Miwa
- Department of Bioengineering, University of California at Los Angeles (UCLA), 90095, USA
| | - Sang Won Han
- Center for Cell Therapy and Molecular, Federal University of São Paulo (UNIFESP), 04044-010, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Brazil
| | - Dino Di Carlo
- Department of Bioengineering, University of California at Los Angeles (UCLA), 90095, USA
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon 21983, Republic of Korea
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 90064, USA; College of Pharmacy, Korea University, 30019, Republic of Korea; Vellore Institute of Technology (VIT), Vellore, 632014, India
| | | | | |
Collapse
|
41
|
Baur E, Tiberghien B, Amstad E. 3D Printing of Double Network Granular Elastomers with Locally Varying Mechanical Properties. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2313189. [PMID: 38530246 DOI: 10.1002/adma.202313189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/22/2024] [Indexed: 03/27/2024]
Abstract
Fast advances in the design of soft actuators and robots demand for new soft materials whose mechanical properties can be changed over short length scales. Elastomers can be formulated as highly stretchable or rather stiff materials and hence, are attractive for these applications. They are most frequently cast such that their composition cannot be changed over short length scales. A method that allows to locally change the composition of elastomers on hundreds of micrometer lengths scales is direct ink writing (DIW). Unfortunately, in the absence of rheomodifiers, most elastomer precursors cannot be printed through DIW. Here, 3D printable double network granular elastomers (DNGEs) whose ultimate tensile strain and stiffness can be varied over an unprecedented range are introduced. The 3D printability of these materials is leveraged to produce an elastomer finger containing rigid bones that are surrounded by a soft skin. Similarly, the rheological properties of the microparticle-based precursors are leveraged to cast elastomer slabs with locally varying stiffnesses that deform and twist in a predefined fashion. These DNGEs are foreseen to open up new avenues in the design of the next generation of smart wearables, strain sensors, prosthesis, soft actuators, and robots.
Collapse
Affiliation(s)
- Eva Baur
- Soft Materials Laboratory, Institute of Materials, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
- National Center of Competence in Research Bio-Inspired Materials, Fribourg, 1700, Switzerland
| | - Benjamin Tiberghien
- Soft Materials Laboratory, Institute of Materials, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Esther Amstad
- Soft Materials Laboratory, Institute of Materials, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
- National Center of Competence in Research Bio-Inspired Materials, Fribourg, 1700, Switzerland
| |
Collapse
|
42
|
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
|
43
|
Richbourg NR, Irakoze N, Kim H, Peyton SR. Outlook and opportunities for engineered environments of breast cancer dormancy. SCIENCE ADVANCES 2024; 10:eadl0165. [PMID: 38457510 PMCID: PMC10923521 DOI: 10.1126/sciadv.adl0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/01/2024] [Indexed: 03/10/2024]
Abstract
Dormant, disseminated breast cancer cells resist treatment and may relapse into malignant metastases after decades of quiescence. Identifying how and why these dormant breast cancer cells are triggered into outgrowth is a key unsolved step in treating latent, metastatic breast cancer. However, our understanding of breast cancer dormancy in vivo is limited by technical challenges and ethical concerns with triggering the activation of dormant breast cancer. In vitro models avoid many of these challenges by simulating breast cancer dormancy and activation in well-controlled, bench-top conditions, creating opportunities for fundamental insights into breast cancer biology that complement what can be achieved through animal and clinical studies. In this review, we address clinical and preclinical approaches to treating breast cancer dormancy, how precisely controlled artificial environments reveal key interactions that regulate breast cancer dormancy, and how future generations of biomaterials could answer further questions about breast cancer dormancy.
Collapse
Affiliation(s)
- Nathan R. Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst Amherst, MA 01003, USA
| |
Collapse
|
44
|
Tuftee C, Alsberg E, Ozbolat IT, Rizwan M. Emerging granular hydrogel bioinks to improve biological function in bioprinted constructs. Trends Biotechnol 2024; 42:339-352. [PMID: 37852853 PMCID: PMC10939978 DOI: 10.1016/j.tibtech.2023.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
Advancements in 3D bioprinting have been hindered by the trade-off between printability and biological functionality. Existing bioinks struggle to meet both requirements simultaneously. However, new types of bioinks composed of densely packed microgels promise to address this challenge. These bioinks possess intrinsic porosity, allowing for cell growth, oxygen and nutrient transport, and better immunomodulatory properties, leading to superior biological functions. In this review, we highlight key trends in the development of these granular bioinks. Using examples, we demonstrate how granular bioinks overcome the trade-off between printability and cell function. Granular bioinks show promise in 3D bioprinting, yet understanding their unique structure-property-function relationships is crucial to fully leverage the transformative capabilities of these new types of bioinks in bioprinting.
Collapse
Affiliation(s)
- Cody Tuftee
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Eben Alsberg
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL 60612, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Orthopedic Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Mechanical & Industrial Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, Chicago, IL 60612, USA
| | - Ibrahim Tarik Ozbolat
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Engineering Science and Mechanics, Penn State University, University Park, PA 16802, USA; Neurosurgery Department, Penn State University; Hershey, PA 17033, USA; Medical Oncology Department, Cukurova University, Adana 01330, Turkey
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA.
| |
Collapse
|
45
|
Sommerfeld IK, Malyaran H, Neuss S, Demco DE, Pich A. Multiresponsive Core-Shell Microgels Functionalized by Nitrilotriacetic Acid. Biomacromolecules 2024; 25:903-923. [PMID: 38170471 DOI: 10.1021/acs.biomac.3c01056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Stimuli-responsive microgels with ionizable functional groups offer versatile applications, e.g., by the uptake of oppositely charged metal ions or guest molecules such as drugs, dyes, or proteins. Furthermore, the incorporation of carboxylic groups enhances mucoadhesive properties, crucial for various drug delivery applications. In this work, we successfully synthesized poly{N-vinylcaprolactam-2,2'-[(5-acrylamido-1-carboxypentyl)azanediyl]diacetic acid} [p(VCL/NTAaa)] microgels containing varying amounts of nitrilotriacetic acid (NTA) using precipitation polymerization. We performed fundamental characterization by infrared (IR) spectroscopy and dynamic and electrophoretic light scattering. Despite their potential multiresponsiveness, prior studies on NTA-functionalized microgels lack in-depth analysis of their stimuli-responsive behavior. This work addresses this gap by assessing the microgel responsiveness to temperature, ionic strength, and pH. Morphological investigations were performed via NMR relaxometry, nanoscale imaging (AFM and SEM), and reaction calorimetry. Finally, we explored the potential application of the microgels by conducting cytocompatibility experiments and demonstrating the immobilization of the model protein cytochrome c in the microgels.
Collapse
Affiliation(s)
- Isabel K Sommerfeld
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
- DWI─Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Hanna Malyaran
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
- Department of Orthodontics, University Hospital of RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
- Institute of Pathology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Dan E Demco
- DWI─Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074 Aachen, Germany
| | - Andrij Pich
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
- DWI─Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, 52074 Aachen, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| |
Collapse
|
46
|
Klar RM, Cox J, Raja N, Lohfeld S. The 3D-McMap Guidelines: Three-Dimensional Multicomposite Microsphere Adaptive Printing. Biomimetics (Basel) 2024; 9:94. [PMID: 38392141 PMCID: PMC10886723 DOI: 10.3390/biomimetics9020094] [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: 12/18/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
Microspheres, synthesized from diverse natural or synthetic polymers, are readily utilized in biomedical tissue engineering to improve the healing of various tissues. Their ability to encapsulate growth factors, therapeutics, and natural biomolecules, which can aid tissue regeneration, makes microspheres invaluable for future clinical therapies. While microsphere-supplemented scaffolds have been investigated, a pure microsphere scaffold with an optimized architecture has been challenging to create via 3D printing methods due to issues that prevent consistent deposition of microsphere-based materials and their ability to maintain the shape of the 3D-printed structure. Utilizing the extrusion printing process, we established a methodology that not only allows the creation of large microsphere scaffolds but also multicomposite matrices into which cells, growth factors, and therapeutics encapsulated in microspheres can be directly deposited during the printing process. Our 3D-McMap method provides some critical guidelines for issues with scaffold shape fidelity during and after printing. Carefully timed breaks, minuscule drying steps, and adjustments to extrusion parameters generated an evenly layered large microsphere scaffold that retained its internal architecture. Such scaffolds are superior to other microsphere-containing scaffolds, as they can release biomolecules in a highly controlled spatiotemporal manner. This capability permits us to study cell responses to the delivered signals to develop scaffolds that precisely modulate new tissue formation.
Collapse
Affiliation(s)
- Roland M Klar
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - James Cox
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Naren Raja
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Stefan Lohfeld
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
47
|
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
|
48
|
Ataie Z, Horchler S, Jaberi A, Koduru SV, El-Mallah JC, Sun M, Kheirabadi S, Kedzierski A, Risbud A, Silva ARAE, Ravnic DJ, Sheikhi A. Accelerating Patterned Vascularization Using Granular Hydrogel Scaffolds and Surgical Micropuncture. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307928. [PMID: 37824280 PMCID: PMC11699544 DOI: 10.1002/smll.202307928] [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: 06/02/2023] [Indexed: 10/14/2023]
Abstract
Bulk hydrogel scaffolds are common in reconstructive surgery. They allow for the staged repair of soft tissue loss by providing a base for revascularization. Unfortunately, they are limited by both slow and random vascularization, which may manifest as treatment failure or suboptimal repair. Rapidly inducing patterned vascularization within biomaterials has profound translational implications for current clinical treatment paradigms and the scaleup of regenerative engineering platforms. To address this long-standing challenge, a novel microsurgical approach and granular hydrogel scaffold (GHS) technology are co-developed to hasten and pattern microvascular network formation. In surgical micropuncture (MP), targeted recipient blood vessels are perforated using a microneedle to accelerate cell extravasation and angiogenic outgrowth. By combining MP with an adjacent GHS with precisely tailored void space architecture, microvascular pattern formation as assessed by density, diameter, length, and intercapillary distance is rapidly guided. This work opens new translational opportunities for microvascular engineering, advancing reconstructive surgery, and regenerative medicine.
Collapse
Affiliation(s)
- Zaman Ataie
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Summer Horchler
- Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Arian Jaberi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Srinivas V Koduru
- Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Jessica C El-Mallah
- Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Mingjie Sun
- Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
| | - Sina Kheirabadi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Alexander Kedzierski
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Aneesh Risbud
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | | | - Dino J Ravnic
- Division of Plastic Surgery, Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Amir Sheikhi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
49
|
Surman F, Asadikorayem M, Weber P, Weber D, Zenobi-Wong M. Ionically annealed zwitterionic microgels for bioprinting of cartilaginous constructs. Biofabrication 2024; 16:025004. [PMID: 38176081 DOI: 10.1088/1758-5090/ad1b1f] [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: 07/21/2023] [Accepted: 01/04/2024] [Indexed: 01/06/2024]
Abstract
Foreign body response (FBR) is a pervasive problem for biomaterials used in tissue engineering. Zwitterionic hydrogels have emerged as an effective solution to this problem, due to their ultra-low fouling properties, which enable them to effectively inhibit FBRin vivo. However, no versatile zwitterionic bioink that allows for high resolution extrusion bioprinting of tissue implants has thus far been reported. In this work, we introduce a simple, novel method for producing zwitterionic microgel bioink, using alginate methacrylate (AlgMA) as crosslinker and mechanical fragmentation as a microgel fabrication method. Photocrosslinked hydrogels made of zwitterionic carboxybetaine acrylamide (CBAA) and sulfobetaine methacrylate (SBMA) are mechanically fragmented through meshes with aperture diameters of 50 and 90µm to produce microgel bioink. The bioinks made with both microgel sizes showed excellent rheological properties and were used for high-resolution printing of objects with overhanging features without requiring a support structure or support bath. The AlgMA crosslinker has a dual role, allowing for both primary photocrosslinking of the bulk hydrogel as well as secondary ionic crosslinking of produced microgels, to quickly stabilize the printed construct in a calcium bath and to produce a microporous scaffold. Scaffolds showed ∼20% porosity, and they supported viability and chondrogenesis of encapsulated human primary chondrocytes. Finally, a meniscus model was bioprinted, to demonstrate the bioink's versatility at printing large, cell-laden constructs which are stable for furtherin vitroculture to promote cartilaginous tissue production. This easy and scalable strategy of producing zwitterionic microgel bioink for high resolution extrusion bioprinting allows for direct cell encapsulation in a microporous scaffold and has potential forin vivobiocompatibility due to the zwitterionic nature of the bioink.
Collapse
Affiliation(s)
- František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, 8032 Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
50
|
Wasnik K, Gupta PS, Mukherjee S, Oviya A, Prakash R, Pareek D, Patra S, Maity S, Rai V, Singh M, Singh G, Yadav DD, Das S, Maiti P, Paik P. Poly( N-acryloylglycine-acrylamide) Hydrogel Mimics the Cellular Microenvironment and Promotes Neurite Growth with Protection from Oxidative Stress. ACS APPLIED BIO MATERIALS 2023; 6:5644-5661. [PMID: 37993284 DOI: 10.1021/acsabm.3c00807] [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: 11/24/2023]
Abstract
In this work, the glycine-based acryloyl monomer is polymerized to obtain a neurogenic polymeric hydrogel for regenerative applications. The synthesized poly(N-acryloylglycine-acrylamide) [poly(NAG-b-A)] nanohydrogel exhibits high swelling (∼1500%) and is mechanically very stable, biocompatible, and proliferative in nature. The poly(NAG-b-A) nanohydrogel provides a stable 3D extracellular mimetic environment and promotes healthy neurite growth for primary cortical neurons by facilitating cellular adhesion, proliferation, actin filament stabilization, and neuronal differentiation. Furthermore, the protective role of the poly(NAG-b-A) hydrogel for the neurons in oxidative stress conditions is revealed and it is found that it is a clinically relevant material for neuronal regenerative applications, such as for promoting nerve regeneration via GSK3β inhibition. This hydrogel additionally plays an important role in modulating the biological microenvironment, either as an agonist and antagonist or as an antioxidant. Furthermore, it favors the physiological responses and eases the neurite growth efficiency. Additionally, we found out that the conversion of glycine-based acryloyl monomers into their corresponding polymer modulates the mechanical performance, mimics the cellular microenvironment, and accelerates the self-healing capability due to the responsive behavior towards reactive oxygen species (ROS). Thus, the p(NAG-b-A) hydrogel could be a potential candidate to induce neuronal regeneration since it provides a physical cue and significantly boosts neurite outgrowth and also maintains the microtubule integrity in neuronal cells.
Collapse
Affiliation(s)
- Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Alagu Oviya
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Ravi Prakash
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Somedutta Maity
- School of Engineering Sciences and Technology, University of Hyderabad, Hyderabad, Telangana State 500 046, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Monika Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Gurmeet Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Desh Deepak Yadav
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Santanu Das
- Department of Ceramic Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Pralay Maiti
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| |
Collapse
|