1
|
Subramaniam V, Abrahan C, Higgins BR, Chisolm SJ, Sweeney B, Duraivel S, Balzano-Nogueira L, Monjure T, Wang CY, Palmer GD, Angelini TE. A functional human liver tissue model: 3D bioprinted co-culture discoids. BIOMATERIALS ADVANCES 2025; 173:214288. [PMID: 40106895 DOI: 10.1016/j.bioadv.2025.214288] [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: 12/11/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
To reduce costs and delays related to developing new and effective drugs, there is a critical need for improved human liver tissue models. Here we describe an approach for 3D bioprinting functional human liver tissue models, in which we fabricate disc-shaped structures (discoids) 200 μm in thickness and 1-3 mm in diameter from mixtures of cells and collagen-1, embedded in a highly permeable support medium made from packed polyethylene glycol (PEG) microgels. We demonstrate that the method is precise, accurate, and scalable; up to 100 tissues/h can be manufactured with a variability and error in diameter of about 4 %. Histologic and immunohistochemical evaluation of printed discs reveal self-organization, cell cohesion, and key liver marker expression. Over the course of three weeks in culture, the tissues stably synthesize albumin and urea at high levels, outperforming spheroid tissue models. We find the tissues express >100 genes associated with molecular absorption, distribution, metabolism, and excretion (ADME) at levels within the range of human liver. The liver tissue models exhibit enzymatic formation of metabolites after exposure to multiple test compounds. Together, these results demonstrate the promise of 3D printed discoids for pharmacological and toxicological applications.
Collapse
Affiliation(s)
- Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Carolina Abrahan
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, FL, United States of America
| | - Brett R Higgins
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States of America
| | - Steven J Chisolm
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Baleigh Sweeney
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Senthilkumar Duraivel
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, United States of America
| | - Leandro Balzano-Nogueira
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States of America
| | - Tia Monjure
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Chih-Yi Wang
- Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America
| | - Glyn D Palmer
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America; Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America; J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
2
|
Zhao W, Yao L, Shen J, Chen S, Zhu S, Chen S, Wang L, Li Y, Liu S, Zhao Q. Advanced Liquid Metal-Based Hydrogels for Flexible Electronics. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27713-27739. [PMID: 40323766 DOI: 10.1021/acsami.5c05225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
With the rapid development of flexible electronics in wearable devices, healthcare devices, and the Internet of Things (IoT), liquid metals (LMs)-based hydrogels have emerged as cutting-edge functional materials due to their high electrical conductivity, tunable mechanical properties, excellent biocompatibility, and unique self-healing properties. Through various physical or chemical methods, LMs can be integrated to form multifunctional LMs-based hydrogels, thus broadening the potential application fields. In this Review, the recent advancement in LMs-based hydrogels for flexible electronics is comprehensively and systematically reviewed from three aspects of synthesis methods, properties, and applications. For the first time, the existing innovative synthesis methods of LMs-based hydrogels are classified and summarized, including patterned LMs on/inside hydrogel substrates, LMs as conductive fillers in polymeric hydrogels, LMs as initiators in hydrogels, and LMs as cross-linkers with organic/inorganic materials. The synthesis mechanism is also stated in detail to highlight the multiple roles of LMs in adjusting the hydrogel properties. The versatile applications of LMs-based hydrogels in flexible electronics, including flexible sensors, wireless communications, electromagnetic interference (EMI) shielding, soft robot actuators, energy storage and conversion, etc., are separately described. Finally, the current challenges and future prospects of LMs-based hydrogels are proposed.
Collapse
Affiliation(s)
- Weiwei Zhao
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Le Yao
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Jiacheng Shen
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Shujiao Chen
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Shujing Zhu
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Shu Chen
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Longlu Wang
- College of Electronic and Optical Engineering & College of Flexible Electronics (Future Technology), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Yang Li
- College of Electronic and Optical Engineering & College of Flexible Electronics (Future Technology), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Shujuan Liu
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Qiang Zhao
- State Key Laboratory of Flexible Electronics (LoFE) and Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
- College of Electronic and Optical Engineering & College of Flexible Electronics (Future Technology), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
3
|
Hua W, Zhang C, Cui H, Mitchell K, Hensley DK, Chen J, Do C, Raymond L, Coulter R, Bandala E, Rubbi F, Chai G, Zhang Z, Liao Y, Zhao D, Wang Y, Gaharwar AK, Jin Y. High-Speed Embedded Ink Writing of Anatomic-Size Organ Constructs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405980. [PMID: 39932855 PMCID: PMC11967790 DOI: 10.1002/advs.202405980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/29/2024] [Indexed: 02/13/2025]
Abstract
Embedded ink writing (EIW) is an emerging 3D printing technique that fabricates complex 3D structures from various biomaterial inks but is limited to a printing speed of ∼10 mm s-1 due to suboptimal rheological properties of particulate-dominated yield-stress fluids when used as liquid baths. In this work, a particle-hydrogel interactive system to design advanced baths with enhanced yield stress and extended thixotropic response time for realizing high-speed EIW is developed. In this system, the interactions between particle additive and three representative polymeric hydrogels enable the resulting nanocomposites to demonstrate different rheological behaviors. Accordingly, the interaction models for the nanocomposites are established, which are subsequently validated by macroscale rheological measurements and advanced microstructure characterization techniques. Filament formation mechanisms in the particle-hydrogel interactive baths are comprehensively investigated at high printing speeds. To demonstrate the effectiveness of the proposed high-speed EIW method, an anatomic-size human kidney construct is successfully printed at 110 mm s-1, which only takes ∼4 h. This work breaks the printing speed barrier in current EIW and propels the maximum printing speed by at least 10 times, providing an efficient and promising solution for organ reconstruction in the future.
Collapse
Affiliation(s)
- Weijian Hua
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Cheng Zhang
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
- State Key Laboratory of High‐Performance Precision ManufacturingDalian University of TechnologyDalianLiaoning116024China
| | - Haoran Cui
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Kellen Mitchell
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Dale K. Hensley
- Center for Nanophase Materials SciencesOak Ridge National LaboratoryOak RidgeTennessee37830USA
| | - Jihua Chen
- Center for Nanophase Materials SciencesOak Ridge National LaboratoryOak RidgeTennessee37830USA
| | - Changwoo Do
- Neutron Scattering DivisionOak Ridge National LaboratoryOak RidgeTennessee37831USA
| | - Lily Raymond
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Ryan Coulter
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Erick Bandala
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Fazlay Rubbi
- Department of Industrial and Manufacturing Systems EngineeringIowa State UniversityAmesIowa50011USA
| | - Guangrui Chai
- Department of OphthalmologyShengjing Hospital of China Medical UniversityShenyangLiaoning110004China
| | - Zhengyi Zhang
- School of Naval Architecture and Ocean EngineeringHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Yiliang Liao
- Department of Industrial and Manufacturing Systems EngineeringIowa State UniversityAmesIowa50011USA
| | - Danyang Zhao
- State Key Laboratory of High‐Performance Precision ManufacturingDalian University of TechnologyDalianLiaoning116024China
| | - Yan Wang
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringTexas A&M University, College StationTexas77843USA
| | - Yifei Jin
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| |
Collapse
|
4
|
Jeon Y, Kim M, Song KH. Development of Hydrogels Fabricated via Stereolithography for Bioengineering Applications. Polymers (Basel) 2025; 17:765. [PMID: 40292646 PMCID: PMC11945500 DOI: 10.3390/polym17060765] [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: 02/19/2025] [Revised: 03/02/2025] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The architectures of hydrogels fabricated with stereolithography (SLA) 3D printing systems have played various roles in bioengineering applications. Typically, the SLA systems successively illuminated light to a layer of photo-crosslinkable hydrogel precursors for the fabrication of hydrogels. These SLA systems can be classified into point-scanning types and digital micromirror device (DMD) types. The point-scanning types form layers of hydrogels by scanning the precursors with a focused light, while DMD types illuminate 2D light patterns to the precursors to form each hydrogel layer at once. Overall, SLA systems were cost-effective and allowed the fabrication of hydrogels with good shape fidelity and uniform mechanical properties. As a result, hydrogel constructs fabricated with the SLA 3D printing systems were used to regenerate tissues and develop lab-on-a-chip devices and native tissue-like models.
Collapse
Affiliation(s)
- Youngjin Jeon
- Department of Nano-Bioengineering, Incheon National University, 119, Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea; (Y.J.); (M.K.)
| | - Minji Kim
- Department of Nano-Bioengineering, Incheon National University, 119, Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea; (Y.J.); (M.K.)
| | - Kwang Hoon Song
- Department of Nano-Bioengineering, Incheon National University, 119, Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea; (Y.J.); (M.K.)
- Research Center of Brain-Machine Interface, Incheon National University, 119, Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| |
Collapse
|
5
|
Bakirci E, Asghari Adib A, Ashraf SF, Feinberg AW. Advancing extrusion-based embedded 3D bioprinting via scientific, engineering, and process innovations. Biofabrication 2025; 17:023002. [PMID: 39965539 DOI: 10.1088/1758-5090/adb7c3] [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: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Extrusion-based embedded 3D bioprinting, where bioinks and biomaterials are extruded within a support bath, has greatly expanded the achievable tissue architectures and complexity of biologic constructs that can be fabricated. However, significant scientific, engineering, and process-related challenges remain to recreate the full anatomic structure and physiologic function required for many therapeutic applications. This perspective explores the future advances in extrusion-based embedded 3D bioprinting that could address these challenges, paving the way for clinical translation of bioprinted tissues.
Collapse
Affiliation(s)
- Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Ali Asghari Adib
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Syed Faaz Ashraf
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
- Department of Materials Science & Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| |
Collapse
|
6
|
Weiss JD, Mermin‐Bunnell A, Solberg FS, Tam T, Rosalia L, Sharir A, Rütsche D, Sinha S, Choi PS, Shibata M, Palagani Y, Nilkant R, Paulvannan K, Ma M, Skylar‐Scott MA. A Low-Cost, Open-Source 3D Printer for Multimaterial and High-Throughput Direct Ink Writing of Soft and Living Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414971. [PMID: 39748617 PMCID: PMC11899504 DOI: 10.1002/adma.202414971] [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: 10/01/2024] [Revised: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Direct ink writing is a 3D printing method that is compatible with a wide range of structural, elastomeric, electronic, and living materials, and it continues to expand its uses into physics, engineering, and biology laboratories. However, the large footprint, closed hardware and software ecosystems, and expense of commercial systems often hamper widespread adoption. This work introduces a compact, low-cost, multimaterial, and high-throughput direct ink writing 3D printer platform with detailed assembly files and instructions provided freely online. In contrast to existing low-cost 3D printers and bioprinters, which typically modify off-the-shelf plastic 3D printers, this system is built from scratch, offering a lower cost and full customizability. Active mixing of cell-laden bioinks, high-throughput production of auxetic lattices using multimaterial multinozzle 3D (MM3D) printing methods, and a high-toughness, photocurable hydrogel for fabrication of heart valves are introduced. Finally, hardware for embedded multinozzle and 3D gradient nozzle printing is developed for producing high-throughput and graded 3D parts. This powerful, simple-to-build, and customizable printing platform can help stimulate a vibrant biomaker community of engineers, biologists, and educators.
Collapse
Affiliation(s)
| | - Alana Mermin‐Bunnell
- Harvard‐MIT Program in Health Science and TechnologyMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Fredrik S. Solberg
- Department of Mechanical EngineeringStanford UniversityStanfordCA94305USA
| | - Tony Tam
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Luca Rosalia
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Amit Sharir
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCA94305USA
| | - Dominic Rütsche
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Soham Sinha
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Perry S. Choi
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCA94305USA
| | - Masafumi Shibata
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCA94305USA
| | - Yellappa Palagani
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCA94305USA
| | - Riya Nilkant
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCA94305USA
| | | | - Michael Ma
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCA94305USA
| | - Mark A. Skylar‐Scott
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- Basic Science and Engineering InitiativeChildren's Heart CenterStanford UniversityStanfordCA94304USA
- Chan Zuckerberg BiohubSan FranciscoCA94158USA
| |
Collapse
|
7
|
Truong H, Abaci A, Gharacheh H, Guvendiren M. Embedded bioprinting of dense cellular constructs in bone allograft-enhanced hydrogel matrices for bone tissue engineering. Biomater Sci 2025. [PMID: 40018866 DOI: 10.1039/d4bm01616e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Bone tissue engineering aims to address critical-sized defects by developing biomimetic scaffolds that promote repair and regeneration. This study introduces a material extrusion-based embedded bioprinting approach to fabricate dense cellular constructs within methacrylated hyaluronic acid (MeHA) hydrogels enhanced with bioactive microparticles. Composite matrices containing human bone allograft or tricalcium phosphate (TCP) particles were evaluated for their rheological, mechanical, and osteoinductive properties. High cell viability (>95%) and uniform strand dimensions were achieved across all bioprinting conditions, demonstrating the method's ability to preserve cellular integrity and structural fidelity. The inclusion of bone or TCP particles did not significantly alter the viscosity, crosslinking kinetics, or compressive modulus of the MeHA hydrogels, ensuring robust mechanical stability and shape retention. However, bone allograft particles significantly enhanced osteogenic differentiation of human mesenchymal stem cells (hMSCs), as evidenced by increased alkaline phosphatase (ALP) activity and calcium deposition. Notably, osteogenesis was observed even in basal media, with a dose-dependent response to bone particle concentration, highlighting the intrinsic bioactivity of allograft particles. This study demonstrates the potential of combining embedded bioprinting with bioactive matrices to create dense, osteoinductive cellular constructs. The ability to induce osteogenesis without external growth factors positions this platform as a scalable and clinically relevant solution for bone repair and regeneration.
Collapse
Affiliation(s)
- Hang Truong
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Alperen Abaci
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Hadis Gharacheh
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| |
Collapse
|
8
|
Aye SSS, Fang Z, Wu MCL, Lim KS, Ju LA. Integrating microfluidics, hydrogels, and 3D bioprinting for personalized vessel-on-a-chip platforms. Biomater Sci 2025; 13:1131-1160. [PMID: 39834160 DOI: 10.1039/d4bm01354a] [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: 01/22/2025]
Abstract
Thrombosis, a major cause of morbidity and mortality worldwide, presents a complex challenge in cardiovascular medicine due to the intricacy of clotting mechanisms in living organisms. Traditional research approaches, including clinical studies and animal models, often yield conflicting results due to the inability to control variables in these complex systems, highlighting the need for more precise investigative tools. This review explores the evolution of in vitro thrombosis models, from conventional polydimethylsiloxane (PDMS)-based microfluidic devices to advanced hydrogel-based systems and cutting-edge 3D bioprinted vascular constructs. We discuss how these emerging technologies, particularly vessel-on-a-chip platforms, are enabling researchers to control previously unmanageable factors, thereby offering unprecedented opportunities to pinpoint specific clotting mechanisms. While PDMS-based devices offer optical transparency and fabrication ease, their inherent limitations, including non-physiological rigidity and surface properties, have driven the development of hydrogel-based systems that better mimic the extracellular matrix of blood vessels. The integration of microfluidics with biomimetic materials and tissue engineering approaches has led to the development of sophisticated models capable of simulating patient-specific vascular geometries, flow dynamics, and cellular interactions under highly controlled conditions. The advent of 3D bioprinting further enables the creation of complex, multi-layered vascular structures with precise spatial control over geometry and cellular composition. Despite significant progress, challenges remain in achieving long-term stability, incorporating immune components, and translating these models to clinical applications. By providing a comprehensive overview of current advancements and future prospects, this review aims to stimulate further innovation in thrombosis research and accelerate the development of more effective, personalized approaches to thrombosis prevention and treatment.
Collapse
Affiliation(s)
- San Seint Seint Aye
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Zhongqi Fang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Mike C L Wu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Khoon S Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
9
|
Cho WW, Park W, Cho DW. Recent trends in embedded 3D bioprinting of vascularized tissue constructs. Biofabrication 2025; 17:022002. [PMID: 39879658 DOI: 10.1088/1758-5090/adafdd] [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: 09/19/2024] [Accepted: 01/29/2025] [Indexed: 01/31/2025]
Abstract
3D bioprinting technology offers significant advantages in the fabrication of tissue and organ structures by allowing precise layer-by-layer patterning of cells and various biomaterials. However, conventional bioinks exhibit poor mechanical properties, which limit their use in the fabrication of large-scale vascularized tissue constructs. To address these limitations, recent studies have focused on the development of rapidly crosslinkable bioinks through chemical modification. These enable rapid crosslinking within minutes, offering substantial advantages for engineering large-scale tissue constructs. Nevertheless, challenges remain in the production of constructs that fully incorporate the complex vascular networks inherent to native tissues. Recently, embedded bioprinting technique, which involves the direct writing of bioink into a support bath that provides physical support, has gained significant attention for enabling the freeform fabrication of 3D structures. This method has been extensively studied and offers the advantage of fabricating structures ranging from tissue constructs with simple vascular channels to complex structures containing multiscale vascular networks. This review presents an overview of the various materials utilized in embedded bioprinting and elucidates the rheological properties of these materials. Furthermore, it examines the current research trends in the biofabrication of vascularized tissue constructs using embedded bioprinting techniques, along with their associated limitations. The review concludes by proposing areas for future improvement, specifically addressing material and biofabrication approaches as well as bioprinting systems.
Collapse
Affiliation(s)
- Won-Woo Cho
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| |
Collapse
|
10
|
Sergis V, Kelly D, Pramanick A, Britchfield G, Mason K, Daly AC. In-situquality monitoring during embedded bioprinting using integrated microscopy and classical computer vision. Biofabrication 2025; 17:025004. [PMID: 39808934 DOI: 10.1088/1758-5090/adaa22] [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: 06/30/2024] [Accepted: 01/14/2025] [Indexed: 01/16/2025]
Abstract
Despite significant advances in bioprinting technology, current hardware platforms lack the capability for process monitoring and quality control. This limitation hampers the translation of the technology into industrial GMP-compliant manufacturing settings. As a key step towards a solution, we developed a novel bioprinting platform integrating a high-resolution camera forin-situmonitoring of extrusion outcomes during embedded bioprinting. Leveraging classical computer vision and image analysis techniques, we then created a custom software module for assessing print quality. This module enables quantitative comparison of printer outputs to input points of the computer-aided design model's 2D projections, measuring area and positional accuracy. To showcase the platform's capabilities, we then investigated compatibility with various bioinks, dyes, and support bath materials for both 2D and 3D print path trajectories. In addition, we performed a detailed study on how the rheological properties of granular support hydrogels impact print quality during embedded bioprinting, illustrating a practical application of the platform. Our results demonstrated that lower viscosity, faster thixotropy recovery, and smaller particle sizes significantly enhance print fidelity. This novel bioprinting platform, equipped with integrated process monitoring, holds great potential for establishing auditable and more reproducible biofabrication processes for industrial applications.
Collapse
Affiliation(s)
- Vasileios Sergis
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, University of Galway, Galway, Ireland
| | - Daniel Kelly
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, University of Galway, Galway, Ireland
| | - Ankita Pramanick
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, University of Galway, Galway, Ireland
| | - Graham Britchfield
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, University of Galway, Galway, Ireland
| | - Karl Mason
- School of Computer Science, University of Galway, University Road, Galway, Ireland
| | - Andrew C Daly
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, University of Galway, Galway, Ireland
| |
Collapse
|
11
|
Kamani KM, Shim YH, Griebler J, Narayanan S, Zhang Q, Leheny RL, Harden JL, Deptula A, Espinosa-Marzal RM, Rogers SA. Linking structural and rheological memory in disordered soft materials. SOFT MATTER 2025; 21:750-759. [PMID: 39791209 DOI: 10.1039/d4sm00953c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Linking the macroscopic flow properties and nanoscopic structure is a fundamental challenge to understanding, predicting, and designing disordered soft materials. Under small stresses, these materials are soft solids, while larger loads can lead to yielding and the acquisition of plastic strain, which adds complexity to the task. In this work, we connect the transient structure and rheological memory of a colloidal gel under cyclic shearing across a range of amplitudes via a generalized memory function using rheo-X-ray photon correlation spectroscopy (rheo-XPCS). Our rheo-XPCS data show that the nanometer scale aggregate-level structure recorrelates whenever the change in recoverable strain over some interval is zero. The macroscopic recoverable strain is therefore a measure of the nano-scale structural memory. We further show that yielding in disordered colloidal materials is strongly heterogeneous and that memories of prior deformation can exist even after the material has been subjected to flow.
Collapse
Affiliation(s)
- Krutarth M Kamani
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| | - Yul Hui Shim
- School of Chemical and Materials Engineering, The University of Suwon, Hwaseong-si, Gyeonggi-do, 18323, Republic of Korea
| | - James Griebler
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| | - Suresh Narayanan
- Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Qingteng Zhang
- Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Robert L Leheny
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - James L Harden
- Department of Physics, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Alexander Deptula
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Illinois, USA, 61801
| | - Rosa M Espinosa-Marzal
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Illinois, USA, 61801
| | - Simon A Rogers
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| |
Collapse
|
12
|
Eom W, Hossain MT, Parasramka V, Kim J, Siu RWY, Sanders KA, Piorkowski D, Lowe A, Koh HG, De Volder MFL, Fudge DS, Ewoldt RH, Tawfick SH. Fast 3D printing of fine, continuous, and soft fibers via embedded solvent exchange. Nat Commun 2025; 16:842. [PMID: 39833187 PMCID: PMC11746892 DOI: 10.1038/s41467-025-55972-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Nature uses fibrous structures for sensing and structural functions as observed in hairs, whiskers, stereocilia, spider silks, and hagfish slime thread skeins. Here, we demonstrate multi-nozzle printing of 3D hair arrays having freeform trajectories at a very high rate, with fiber diameters as fine as 1.5 µm, continuous lengths reaching tens of centimeters, and a wide range of materials with elastic moduli from 5 MPa to 3500 MPa. This is achieved via 3D printing by rapid solvent exchange in high yield stress micro granular gel, leading to radial solidification of the extruded polymer filament at a rate of 2.33 μm/s. This process extrudes filaments at 5 mm/s, which is 500,000 times faster than meniscus printing owing to the rapid solidification which prevents capillarity-induced fiber breakage. This study demonstrates the potential of 3D printing by rapid solvent exchange as a fast and scalable process for replicating natural fibrous structures for use in biomimetic functions.
Collapse
Affiliation(s)
- Wonsik Eom
- Department of Fiber Convergence Material Engineering, Dankook University, Yongin-si, Republic of Korea
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Mohammad Tanver Hossain
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Vidush Parasramka
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jeongmin Kim
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Ryan W Y Siu
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Kate A Sanders
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Dakota Piorkowski
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | - Andrew Lowe
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | - Hyun Gi Koh
- Department of Biological and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | | | - Douglas S Fudge
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA
| | - Randy H Ewoldt
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Sameh H Tawfick
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Engineering, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Lyu Y, Ji Z, Liu D, Xu X, Guo R, Shi X, Wang X. Spider-silk inspired ultrafast alkali-induced molecular aggregation for 3D printing arbitrary tubular hydrogels. MATERIALS HORIZONS 2025; 12:520-530. [PMID: 39494672 DOI: 10.1039/d4mh01291g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Fabricating tubular hydrogel models with arbitrary structural complexity and controllable diameters using an ultrafast, facile yet universal method is desirable for vascular prototypes yet still a great challenge. Herein, inspired by the denaturing ability of spider silks, a novel strategy to induce complexation via applying highly concentrated alkali into a polyvinyl alcohol/ionic liquid (PVA/IL) solution, i.e., alkali-induced molecular aggregation (AMA), is proposed to achieve such purpose. This strategy enables the rapid and facile fabrication of tubular hydrogel architectures with tunable diameters, controllable thicknesses, and excellent mechanical performance with a tensile strength of up to 1.1 MPa and stretchability exceeding 600%. Importantly, this novel strategy combined with 3D printing facilitates the rapid fabrication of a variety of precise tubular hydrogel models with connected cavity structures which are difficult to achieve using current methods. This ultrafast solidification strategy could also be extended to various alkalis, cations and anions to build different hydrogels, showcasing its versatility and universality. Hence, this strategy can be pioneering to rapidly fabricate complex three-dimensional and hollow enclosed hydrogel models for simulating endovascular interventional therapy.
Collapse
Affiliation(s)
- Yang Lyu
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai Zhongke Research Institute of Advanced Materials and Green Chemical Engineering, Yantai, 264006, China
| | - Zhongying Ji
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai Zhongke Research Institute of Advanced Materials and Green Chemical Engineering, Yantai, 264006, China
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China.
| | - Di Liu
- Key Laboratory of Rubber-plastics, Ministry of Education, School of Polymer Science and Engineering, Qingdao University of Science & Technology, Qingdao, 266042, China
| | - Xinqiang Xu
- School of Chemistry and Chemical Engineering, Key Laboratory of Materials-Oriented Chemical Engineering of Xinjiang Uygur Autonomous Region, Shihezi University, Shihezi, 832003, China
| | - Rui Guo
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai Zhongke Research Institute of Advanced Materials and Green Chemical Engineering, Yantai, 264006, China
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China.
| | - Xinyan Shi
- Key Laboratory of Rubber-plastics, Ministry of Education, School of Polymer Science and Engineering, Qingdao University of Science & Technology, Qingdao, 266042, China
| | - Xiaolong Wang
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai Zhongke Research Institute of Advanced Materials and Green Chemical Engineering, Yantai, 264006, China
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China.
| |
Collapse
|
14
|
Chen S, Tan L, Serpooshan V, Chen H. A 3D bioprinted adhesive tissue engineering scaffold to repair ischemic heart injury. Biomater Sci 2025; 13:506-522. [PMID: 39639799 DOI: 10.1039/d4bm00988f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Adhesive tissue engineering scaffold (ATES) devices can be secured on tissues by relying on their intrinsic adhesive properties, hence, avoiding the complications such as host tissue/scaffold damage that are associated with conventional scaffold fixation methods like suturing or bioglue. This study introduces a new generation of three-dimensional (3D) bioprinted ATES systems for use as cardiac patches to regenerate the adult human heart. Tyramine-modified methacrylated hyaluronic acid (HAMA-tyr), gelatin methacrylate (GelMA), and gelatin were used to create the hybrid bioink formulation with self-adhesive properties. ATESs were bioprinted and further modified to improve the adhesion properties. In-depth characterization of printing fidelity, pore size, mechanical properties, swelling behavior, as well as biocompatibility was used to create ATESs with optimal biological function. Following in vitro testing, the ATESs were tested in a mouse model of myocardial infarction to study the scaffold adhesive strength in biological milieu. The method developed in this study can be used to manufacture off-the-shelf ATESs with complex cellular and extracellular architecture, with robust potential for clinical translation into a variety of personalized tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Lindan Tan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| |
Collapse
|
15
|
Caiado Decarli M, Ferreira HP, Sobreiro‐Almeida R, Teixeira FC, Correia TR, Babilotte J, Olijve J, Custódio CA, Gonçalves IC, Mota C, Mano JF, Moroni L. Embedding Bioprinting of Low Viscous, Photopolymerizable Blood-Based Bioinks in a Crystal Self-Healing Transparent Supporting Bath. SMALL METHODS 2025; 9:e2400857. [PMID: 38970553 PMCID: PMC11740956 DOI: 10.1002/smtd.202400857] [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: 06/08/2024] [Indexed: 07/08/2024]
Abstract
Protein-based hydrogels have great potential to be used as bioinks for biofabrication-driven tissue regeneration strategies due to their innate bioactivity. Nevertheless, their use as bioinks in conventional 3D bioprinting is impaired due to their intrinsic low viscosity. Using embedding bioprinting, a liquid bioink is printed within a support that physically holds the patterned filament. Inspired by the recognized microencapsulation technique complex coacervation, crystal self-healing embedding bioprinting (CLADDING) is introduced based on a highly transparent crystal supporting bath. The suitability of distinct classes of gelatins is evaluated (i.e., molecular weight distribution, isoelectric point, and ionic content), as well as the formation of gelatin-gum arabic microparticles as a function of pH, temperature, solvent, and mass ratios. Characterizing and controlling this parametric window resulted in high yields of support bath with ideal self-healing properties for interaction with protein-based bioinks. This support bath achieved transparency, which boosted light permeation within the bath. Bioprinted constructs fully composed of platelet lysates encapsulating a co-culture of human mesenchymal stromal cells and endothelial cells are obtained, demonstrating a high-dense cellular network with excellent cell viability and stability over a month. CLADDING broadens the spectrum of photocrosslinkable materials with extremely low viscosity that can now be bioprinted with sensitive cells without any additional support.
Collapse
Affiliation(s)
- Monize Caiado Decarli
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Biomaterials & Biomedical TechnologyUniversity Medical Center Groningen/University of GroningenA. Deusinglaan 1GroningenAV 9713The Netherlands
| | - Helena P. Ferreira
- i3S – Instituto de Investigação e Inovação em Saúde/INEB – Instituto de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen 208Porto4200‐180Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel SalazarUniversidade do PortoPorto4050‐313Portugal
| | - Rita Sobreiro‐Almeida
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Filipa C. Teixeira
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Tiago R. Correia
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Joanna Babilotte
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Jos Olijve
- Rousselot BiomedicalExpertise CenterMeulestedekaai 81Ghent9000Belgium
| | - Catarina A. Custódio
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
- Metatissue, PCICreative Science Park Aveiro RegionVia do ConhecimentoÍlhavo3830‐352Portugal
| | - Inês C. Gonçalves
- i3S – Instituto de Investigação e Inovação em Saúde/INEB – Instituto de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen 208Porto4200‐180Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel SalazarUniversidade do PortoPorto4050‐313Portugal
| | - Carlos Mota
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - João F. Mano
- CICECO – Department of ChemistryAveiro Institute of MaterialsUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|
16
|
Khaled Wassif R, Daihom BA, Maniruzzaman M. FRESH 3D printing of zoledronic acid-loaded chitosan/alginate/hydroxyapatite composite thermosensitive hydrogel for promoting bone regeneration. Int J Pharm 2024; 667:124898. [PMID: 39500473 DOI: 10.1016/j.ijpharm.2024.124898] [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: 08/26/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/09/2024]
Abstract
The aim of this study was to develop a composite thermosensitive hydrogel for bone regeneration applications. This hydrogel consisted of chitosan, alginate and hydroxyapatite, and was loaded with zoledronic acid as a model drug. The feasibility of three-dimensional (3D) printing of the thermosensitive hydrogel using the extrusion based technique was investigated. The 3D printing technique called Freeform Reversible Embedded Suspended Hydrogel (FRESH) printing was employed for this purpose. To characterize the composite hydrogels, several tests were conducted. The gelation time, rheological properties, and in vitro drug release were analyzed. Additionally, the cell viability test on human osteosarcoma MG-63 cells for the composite hydrogel was assessed using an MTT assay. The results of the study showed that the zoledronic acid-loaded composite thermosensitive hydrogel was successfully printed using the FRESH 3D printing technique which was not possible otherwise i.e., by using traditional 3D printing techniques. Further examination of the printed constructs using a Scanning Electron Microscope revealed the presence of porous and layered structures. The gelation times of the composite thermosensitive hydrogel was determined to be 10 and 20 min, respectively for scaffolds with and without HA, indicating the successful formation of the gel within a reasonable time to the FRESH technique. The flow behavior of the hydrogel was found to be pseudoplastic, following a non-Newtonian flow pattern with Farrow's constant (N) values of 1.708 and 1.853 for scaffolds with and without hydroxyapatite, respectively. In terms of drug release, scaffolds prepared with and without hydroxyapatite reached nearly 100% of zoledronic acid release in 360 h and 48 h, respectively. The cell viability test on human osteosarcoma MG-63 cells using MTT assay has shown increased cell viability % in the case of composite hydrogel, indicating biocompatibility of the scaffold. Overall, this study successfully developed a composite thermosensitive hydrogel loaded with zoledronic acid for bone regeneration applications and was 3D printed using the FRESH 3D printing technique. The results of this study provide valuable insights into the potential use of this composite hydrogel for future biomedical applications.
Collapse
Affiliation(s)
- Reem Khaled Wassif
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Baher A Daihom
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohammed Maniruzzaman
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
17
|
Rosas J, Campanale JP, Harwood JL, Li L, Bae R, Cheng S, Tsou JM, Kaiser KM, Engle DD, Montell DJ, Pitenis AA. Differential Effects of Confinement on the Dynamics of Normal and Tumor-Derived Pancreatic Ductal Organoids. ACS APPLIED BIO MATERIALS 2024; 7:8489-8502. [PMID: 39576883 PMCID: PMC11653396 DOI: 10.1021/acsabm.4c01301] [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: 09/09/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a cancer of the epithelia comprising the ductal network of the pancreas. During disease progression, PDAC tumors recruit fibroblasts that promote fibrosis, increasing local tissue stiffness and subjecting epithelial cells to increased compressive forces. Previous in vitro studies have documented cytoskeletal and nuclear adaptation following compressive stresses in two-dimensional (2D) and three-dimensional (3D) environments. However, a comparison of the responses of normal and tumor-derived ductal epithelia to physiologically relevant confinement remains underexplored, especially in 3D organoids. Here we control confinement with an engineered 3D microenvironment composed of Matrigel mixed with a low yield stress granular microgel. Normal and tumor-derived murine pancreas organoids (normal and tumor) were cultured for 48 h within this composite 3D environment or in pure Matrigel to investigate the effects of confinement on morphogenesis and lumen expansion. In confinement, tumor organoids (mT) formed a lumen that expanded rapidly, whereas normal organoids (mN) expanded more slowly. Moreover, a majority of normal organoids in more-confined conditions exhibited an inverted apicobasal polarity compared to those in less-confined conditions. Tumor organoids exhibited a collective "pulsing" behavior that increased in confinement. These pulses generated forces sufficient to locally overcome the yield stress of the microgels in the direction of organoid expansion. Normal organoids more commonly exhibit unidirectional rotation. Our in vitro microgel confinement platform enabled the discovery of two distinct modes of collective force generation in organoids that may shed light on the mutual interactions between tumors and the microenvironment. These insights into in vitro dynamics may deepen our understanding of how the confinement of healthy cells within a fibrotic tumor niche disrupts tissue organization and function in vivo.
Collapse
Affiliation(s)
- Jonah
M. Rosas
- Department
of Biomolecular Science & Engineering Program, University of California, Santa
Barbara, California 93106, United States
| | - Joseph P. Campanale
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Jacob L. Harwood
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Lufei Li
- Department
of Statistics and Applied Probability, University
of California, Santa Barbara, California 93106, United States
| | - Rachel Bae
- Department
of Chemistry & Biochemistry, University
of California, Santa Barbara, California 93106, United States
- Materials
Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Shujun Cheng
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Julia M. Tsou
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Kathi M. Kaiser
- Department
of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Materials
Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Dannielle D. Engle
- Salk Institute
for Biological Studies, La Jolla, California 92037, United States
| | - Denise J. Montell
- Department
of Biomolecular Science & Engineering Program, University of California, Santa
Barbara, California 93106, United States
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Angela A. Pitenis
- Materials
Department, University of California, Santa Barbara, California 93106, United States
- Materials
Research Laboratory, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
18
|
Claxton N, Luse MA, Isakson BE, Highley CB. Engineering Granular Hydrogels without Interparticle Cross-Linking to Support Multicellular Organization. ACS Biomater Sci Eng 2024; 10:7594-7605. [PMID: 39585331 PMCID: PMC11632665 DOI: 10.1021/acsbiomaterials.4c01563] [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: 08/26/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Advancing three-dimensional (3D) tissue constructs is central to creating in vitro models and engineered tissues that recapitulate biology. Materials that are permissive to cellular behaviors, including proliferation, morphogenesis of multicellular structures, and motility, will support the emergence of tissue structures. Granular hydrogels in which there is no interparticle cross-linking exhibit dynamic properties that may be permissive to such cellular behaviors. However, designing granular hydrogels that lack interparticle cross-linking but support cellular self-organization remains underexplored relative to granular systems stabilized by interparticle cross-linking. In this study, we developed a polyethylene glycol-based granular hydrogel system, with average particle diameters under 40 μm. This granular hydrogel exhibited bulk stress-relaxing behaviors and compatibility with custom microdevices to sustain cell cultures without degradation. The system was studied in conjunction with cocultures of endothelial cells and fibroblasts, known for their spontaneous network formation. Cross-linking, porosity, and cell-adhesive ligands (such as RGD) were manipulated to control system properties. Toward supporting cellular activity, increased porosity was found to enhance the formation of cellular networks, whereas RGD reduced network formation in the system studied. This research highlights the potential of un-cross-linked granular systems to support morphogenetic processes, like vasculogenesis and tissue maturation, offering insights into material design for 3D cell culture systems.
Collapse
Affiliation(s)
- Natasha
L. Claxton
- Department
of Biomedical Engineering, University of
Virginia, Charlottesville, Virginia 22903, United States
| | - Melissa A. Luse
- Department
of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
| | - Brant E. Isakson
- Department
of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
- Robert
M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
| | - Christopher B. Highley
- Department
of Biomedical Engineering, University of
Virginia, Charlottesville, Virginia 22903, United States
- Department
of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| |
Collapse
|
19
|
Jiang Z, Jin B, Liang Z, Wang Y, Ren S, Huang Y, Li C, Sun H, Li Y, Liu L, Li N, Wang J, Cui Z, Huang P, Yang H, Mao Y, Ye H. Liver bioprinting within a novel support medium with functionalized spheroids, hepatic vein structures, and enhanced post-transplantation vascularization. Biomaterials 2024; 311:122681. [PMID: 38944968 DOI: 10.1016/j.biomaterials.2024.122681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/28/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Cell-laden bioprinting is a promising biofabrication strategy for regenerating bioactive transplants to address organ donor shortages. However, there has been little success in reproducing transplantable artificial organs with multiple distinctive cell types and physiologically relevant architecture. In this study, an omnidirectional printing embedded network (OPEN) is presented as a support medium for embedded 3D printing. The medium is state-of-the-art due to its one-step preparation, fast removal, and versatile ink compatibility. To test the feasibility of OPEN, exceptional primary mouse hepatocytes (PMHs) and endothelial cell line-C166, were used to print hepatospheroid-encapsulated-artificial livers (HEALs) with vein structures following predesigned anatomy-based printing paths in OPEN. PMHs self-organized into hepatocyte spheroids within the ink matrix, whereas the entire cross-linked structure remained intact for a minimum of ten days of cultivation. Cultivated HEALs maintained mature hepatic functions and marker gene expression at a higher level than conventional 2D and 3D conditions in vitro. HEALs with C166-laden vein structures promoted endogenous neovascularization in vivo compared with hepatospheroid-only liver prints within two weeks of transplantation. Collectively, the proposed platform enables the manufacture of bioactive tissues or organs resembling anatomical architecture, and has broad implications for liver function replacement in clinical applications.
Collapse
Affiliation(s)
- Zhuoran Jiang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China; Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK; Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Yinhan Wang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Shuai Ren
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Yongfa Huang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Changcan Li
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Hang Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yunzhu Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Li Liu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Nianlin Li
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Jinzhuo Wang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK; The Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou, 215123, China
| | - Pengyu Huang
- Engineering Research Center of Pulmonary and Critical Care Technology and Device (MOE of China), Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK; The Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou, 215123, China.
| |
Collapse
|
20
|
Lee MC, Jodat YA, Endo Y, Rodríguez-delaRosa A, Zhang T, Karvar M, Tanoury ZA, Quint J, Kamperman T, Kiaee K, Ochoa SL, Shi K, Huang Y, Rosales MP, Lee H, Kim J, Ceron EL, Reyes IG, Panayi AC, Wang X, Kim KT, Moon JI, Park SG, Lee K, Calabrese MA, Lee J, Tamayol A, Lee L, Pourquié O, Kim WJ, Sinha I, Shin SR. Engineering large-scale hiPSC-derived vessel-integrated muscle-like lattices for enhanced volumetric muscle regeneration. Trends Biotechnol 2024; 42:1715-1744. [PMID: 39306493 PMCID: PMC11625013 DOI: 10.1016/j.tibtech.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 12/08/2024]
Abstract
Engineering biomimetic tissue implants with human induced pluripotent stem cells (hiPSCs) holds promise for repairing volumetric tissue loss. However, these implants face challenges in regenerative capability, survival, and geometric scalability at large-scale injury sites. Here, we present scalable vessel-integrated muscle-like lattices (VMLs), containing dense and aligned hiPSC-derived myofibers alongside passively perfusable vessel-like microchannels inside an endomysium-like supporting matrix using an embedded multimaterial bioprinting technology. The contractile and millimeter-long myofibers are created in mechanically tailored and nanofibrous extracellular matrix-based hydrogels. Incorporating vessel-like lattice enhances myofiber maturation in vitro and guides host vessel invasion in vivo, improving implant integration. Consequently, we demonstrate successful de novo muscle formation and muscle function restoration through a combinatorial effect between improved graft-host integration and its increased release of paracrine factors within volumetric muscle loss injury models. The proposed modular bioprinting technology enables scaling up to centimeter-sized prevascularized hiPSC-derived muscle tissues with custom geometries for next-generation muscle regenerative therapies.
Collapse
Affiliation(s)
- Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Medicinal Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792 Republic of Korea
| | - Yasamin A. Jodat
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yori Endo
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Ting Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Mehran Karvar
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Tom Kamperman
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Sofia Lara Ochoa
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kun Shi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Montserrat Pineda Rosales
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Hyeseon Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Jiseong Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Eder Luna Ceron
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Isaac Garcia Reyes
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Adriana C. Panayi
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xichi Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ki-Tae Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae-I Moon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Gwa Park
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kangju Lee
- Department of Healthcare and Medical Engineering, Chonnam National University, Yeosu 59626, South Korea
| | - Michelle A. Calabrese
- Chemical Engineering and Materials Science Department, University of Minnesota, Minneapolis, MN 55455, USA
| | - Junmin Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Luke Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Woo-Jin Kim
- Correspondence: (I.S.), (W.J.K.), (S.R.S.), Twitter: Yasamin A. Jodat: @YasaminJodat
| | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Lead contact
| |
Collapse
|
21
|
Deng X, Qi C, Meng S, Dong H, Wang T, Liu Z, Kong T. All-Aqueous Embedded 3D Printing for Freeform Fabrication of Biomimetic 3D Constructs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406825. [PMID: 39520386 DOI: 10.1002/adma.202406825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
All-aqueous embedded 3D printing, which involves extruding inks in an aqueous bath, has emerged as a transformative platform for the freeform fabrication of 3D constructs with precise control. The use of a supporting bath not only enables the printing of arbitrarily designed 3D constructs but also broadens ink selection for various soft matters, advancing the wide application of this technology. This review focuses on recent progress in the freeform preparation of 3D constructs using all-aqueous embedded 3D printing. It begins by discussing the significance of ultralow interfacial tension in all-liquid embedded printing and highlights the fundamental concepts and properties of all-aqueous system. The review then introduces recent advances in all-aqueous embedded 3D printing and clarifies the key factors affecting printing stability and shape fidelity, aiming to guide expansion and assessment of emerging printing systems used for various representative applications. Furthermore, it proposes the potential scope and applications of this technology, including in vitro models, cytomimetic microreactors, and soft ionic electronics. Finally, the review discusses the challenges facing current all-aqueous embedded 3D printing and offers future perspectives on possible improvements and developments.
Collapse
Affiliation(s)
- Xiaokang Deng
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, 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
| | - Cheng Qi
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Si Meng
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Haifeng Dong
- Huizhou Institute of Green Energy and Advanced Materials, Huizhou, Guangdong, 516081, China
| | - Tianfu Wang
- 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
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Tiantian Kong
- 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
| |
Collapse
|
22
|
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
|
23
|
Rector Iv JA, McBride L, Weber CM, Grossman K, Sorets A, Ventura-Antunes L, Holtz I, Young K, Schrag M, Lippmann ES, Bellan LM. Fabrication of endothelialized capillary-like microchannel networks using sacrificial thermoresponsive microfibers. Biofabrication 2024; 17:015023. [PMID: 39401530 PMCID: PMC11575475 DOI: 10.1088/1758-5090/ad867d] [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: 02/22/2024] [Revised: 07/28/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024]
Abstract
In the body, capillary beds fulfill the metabolic needs of cells by acting as the sites of diffusive transport for vital gasses and nutrients. In artificial tissues, replicating the scale and complexity of capillaries has proved challenging, especially in a three-dimensional context. In order to better develop thick artificial tissues, it will be necessary to recreate both the form and function of capillaries. Here we demonstrate a top-down method of patterning hydrogels using sacrificial templates formed from thermoresponsive microfibers whose size and architecture approach those of natural capillaries. Within the resulting microchannels, we cultured endothelial monolayers that remain viable for over three weeks and exhibited functional barrier properties. Additionally, we cultured endothelialized microchannels within hydrogels containing fibroblasts and characterized the viability of the co-cultures to demonstrate this approach's potential when applied to cell-laden hydrogels. This method represents a step forward in the evolution of artificial tissues and a path towards producing viable capillary-scale microvasculature for engineered organs.
Collapse
Affiliation(s)
- John A Rector Iv
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Lucas McBride
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Callie M Weber
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Kira Grossman
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Alexander Sorets
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Lissa Ventura-Antunes
- School of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Isabella Holtz
- Department of Cognitive Studies, Vanderbilt University, Nashville, TN, United States of America
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, TN, United States of America
| | - Katherine Young
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Matthew Schrag
- School of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Ethan S Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Leon M Bellan
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
24
|
Friedrich LM, Gunther RT. Simulated inter-filament fusion in embedded 3D printing. Biofabrication 2024; 17:015022. [PMID: 39509819 DOI: 10.1088/1758-5090/ad8fd5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 11/07/2024] [Indexed: 11/15/2024]
Abstract
In embedded 3D printing (EMB3D), a nozzle extrudes continuous filaments inside of a viscoelastic support bath. Compared to other extrusion processes, EMB3D enables softer structures and print paths that conform better to the shape of the part, allowing for complex structures such as tissues and organs. However, strategies for high-quality dimensional accuracy and mechanical properties remain undocumented in EMB3D. This work uses computational fluid dynamics simulations in OpenFOAM to probe the underlying physics behind two processes: deformation of the printed part due to nearby nozzle motion and fusion between neighboring filaments during printing. Through simulations, we disentangle yielding from viscous dissipation, and we isolate interfacial tension effects from rheology effects, which are difficult to separate in experiments. Critically, these simulations find that disturbance and fusion are controlled by the flow of support fluid around the nozzle. To avoid part deformation, the nozzle must remain far from existing parts during non-printing moves, moreso when traveling next to the part than above the part and especially when the interfacial tension between the ink and support is non-zero. Additionally, because support can become trapped between filaments at zero interfacial tension, the spacing between filaments must be tight enough to produce over-printing, or printing too much material for the designed space. In non-Newtonian fluids, spacings for vertical walls must be even tighter than spacings for horizontal planes. At these spacings, printing a new filament sometimes creates and sometimes mitigates shape defects in the old filament. While non-zero ink-support interfacial tensions produce better inter-filament fusion than zero interfacial tension, interfacial tension also produces shape defects. Slicing algorithms that consider these unique EMB3D defects are needed to improve mechanical properties and dimensional accuracy of bioprinted constructs.
Collapse
Affiliation(s)
- Leanne M Friedrich
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899, United States of America
| | - Ross T Gunther
- Materials Science and Engineering, Georgia Institute of Technology, North Avenue, GA, Atlanta 30332, United States of America
| |
Collapse
|
25
|
Xu Y, Shen Y. The Assembly of Miniaturized Droplets toward Functional Architectures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404366. [PMID: 39380419 DOI: 10.1002/smll.202404366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/19/2024] [Indexed: 10/10/2024]
Abstract
Recent explorations of bioengineering have generated new concepts and strategies for the processing of soft and functional materials. Droplet assembly techniques can address problems in the construction of extremely soft architectures by expanding the manufacturing capabilities using droplets containing liquid or hydrogels including weak hydrogels. This Perspective sets out to provide a brief overview of this growing field, and discusses the challenges and opportunities ahead. The study highlights the recent key advances of materials and architectures from hitherto effective droplet-assembly technologies, as well as the applications in biomedical and bioengineering fields from artificial tissues to bioreactors. It is envisaged that these assembled architectures, as nature-inspired models, will stimulate the discovery of biomaterials and miniaturized platforms for interdisciplinary research in health, biotechnology, and sustainability.
Collapse
Affiliation(s)
- Yufan Xu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Yi Shen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
26
|
Nakamura K, Di Caprio N, Burdick JA. Engineered Shape-Morphing Transitions in Hydrogels Through Suspension Bath Printing of Temperature-Responsive Granular Hydrogel Inks. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410661. [PMID: 39358935 PMCID: PMC11588557 DOI: 10.1002/adma.202410661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/12/2024] [Indexed: 10/04/2024]
Abstract
4D printing of hydrogels is an emerging technology used to fabricate shape-morphing soft materials that are responsive to external stimuli for use in soft robotics and biomedical applications. Soft materials are technically challenging to process with current 4D printing methods, which limits the design and actuation potential of printed structures. Here, a simple multi-material 4D printing technique is developed that combines dynamic temperature-responsive granular hydrogel inks based on hyaluronic acid, whose actuation is modulated via poly(N-isopropylacrylamide) crosslinker design, with granular suspension bath printing that provides structural support during and after the printing process. Granular hydrogels are easily extruded upon jamming due to their shear-thinning properties and their porous structure enables rapid actuation kinetics (i.e., seconds). Granular suspension baths support responsive ink deposition into complex patterns due to shear-yielding to fabricate multi-material objects that can be post-crosslinked to obtain anisotropic shape transformations. Dynamic actuation is explored by varying printing patterns and bath shapes, achieving complex shape transformations such as 'S'-shaped and hemisphere structures. Furthermore, stepwise actuation is programmed into multi-material structures by using microgels with varied transition temperatures. Overall, this approach offers a simple method to fabricate programmable soft actuators with rapid kinetics and precise control over shape morphing.
Collapse
Affiliation(s)
- Keisuke Nakamura
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
| | - Nikolas Di Caprio
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Jason A. Burdick
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303 USA
| |
Collapse
|
27
|
Li S, Li J, Xu J, Shen Y, Shang X, Li H, Wang J, Liu Y, Qiang L, Qiao Z, Wang J, He Y, Hu Y. Removal-Free and Multicellular Suspension Bath-Based 3D Bioprinting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406891. [PMID: 39394784 DOI: 10.1002/adma.202406891] [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: 05/14/2024] [Revised: 09/27/2024] [Indexed: 10/14/2024]
Abstract
Suspension bath-based 3D bioprinting (SUB3BP) is effective in creating engineered vascular structures. The transfer of oxygen and nutrients via engineered vascular networks is necessary for tissue or organ survival and integration following transplantation. Existing SUB3BP techniques face challenges in fabricating hierarchical structures with multicellular organization, including issues related to suspension bath removal, restricted material choices, and low accuracy. A next-generation SUB3BP technique that is removal-free and multicellular is presented. A simple, storable, stable, and scalable starch hydrogel design leverages the diverse spectrum of hydrogels available for use in SUB3BP. Starch granules (8.1 µm) create vascular structures with minimal surface roughness (2.5 µm) that simulate more natural vessel walls compared to prior research. The development of cells and organoids, as well as the bioprinting of multicellular skin models with vasculature, demonstrates that starch suspension baths eliminate the removal process and have the potential for fabricating artificial tissue with a hierarchical structure and multicellular distribution.
Collapse
Affiliation(s)
- Shuai Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jianping Li
- Zhejiang Key Laboratory of Precision Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jian Xu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifan Shen
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiushuai Shang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hangyu Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jingwen Wang
- Zhejiang Key Laboratory of Precision Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yihao Liu
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lei Qiang
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhiguang Qiao
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jinwu Wang
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yong He
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
28
|
Vialetto J, Ramakrishna SN, Isa L, Laurati M. Effect of particle stiffness and surface properties on the non-linear viscoelasticity of dense microgel suspensions. J Colloid Interface Sci 2024; 672:814-823. [PMID: 38878623 DOI: 10.1016/j.jcis.2024.05.214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 07/07/2024]
Abstract
HYPOTHESIS Particle surface chemistry and internal softness are two fundamental parameters in governing the mechanical properties of dense colloidal suspensions, dictating structure and flow, therefore of interest from materials fabrication to processing. EXPERIMENTS Here, we modulate softness by tuning the crosslinker content of poly(N-isopropylacrylamide) microgels, and we adjust their surface properties by co-polymerization with polyethylene glycol chains, controlling adhesion, friction and fuzziness. We investigate the distinct effects of these parameters on the entire mechanical response from restructuring to complete fluidization of jammed samples at varying packing fractions under large-amplitude oscillatory shear experiments, and we complement rheological data with colloidal-probe atomic force microscopy to unravel variations in the particles' surface properties. FINDINGS Our results indicate that surface properties play a fundamental role at smaller packings; decreasing adhesion and friction at contact causes the samples to yield and fluidify in a lower deformation range. Instead, increasing softness or fuzziness has a similar effect at ultra-high densities, making suspensions able to better adapt to the applied shear and reach complete fluidization over a larger deformation range. These findings shed new light on the single-particle parameters governing the mechanical response of dense suspensions subjected to deformation, offering synthetic approaches to design materials with tailored mechanical properties.
Collapse
Affiliation(s)
- Jacopo Vialetto
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino (FI), Italy; Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland; Consorzio interuniversitario per lo sviluppo dei Sistemi a Grande Interfase (CSGI), via della Lastruccia 3, 50019 Sesto Fiorentino (FI), Italy.
| | - Shivaprakash N Ramakrishna
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland
| | - Lucio Isa
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093 Zürich, Switzerland
| | - Marco Laurati
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino (FI), Italy; Consorzio interuniversitario per lo sviluppo dei Sistemi a Grande Interfase (CSGI), via della Lastruccia 3, 50019 Sesto Fiorentino (FI), Italy.
| |
Collapse
|
29
|
Friedrich LM, Woodcock JW. Filament Disturbance and Fusion during Embedded 3D Printing of Silicones. ACS Biomater Sci Eng 2024; 10:6690-6710. [PMID: 39235368 DOI: 10.1021/acsbiomaterials.4c01014] [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: 09/06/2024]
Abstract
Embedded 3D printing (EMB3D) is an additive manufacturing technique that enables complex fabrication of soft materials including tissues and silicones. In EMB3D, a nozzle writes continuous filaments into a support bath consisting of a yield stress fluid. Lack of fusion defects between filaments can occur because the nozzle pushes support fluid into existing filaments, preventing coalescence. Interfacial tension was previously proposed as a tool to drive interfilament fusion. However, interfacial tension can also drive rupture and shrinkage of printed filaments. Here, we evaluate the efficacy of interfacial tension as a tool to control defects in EMB3D. Using polydimethylsiloxane (PDMS)-based inks with varying amounts of fumed silica and surfactant, printed into Laponite in water supports, we evaluate the effect of rheology, interfacial tension, print speeds, and interfilament spacings on defects. We print pairs of parallel filaments at varying orientations in the bath and use digital image analysis to quantify shrinkage, rupture, fusion, and positioning defects. By comparing disturbed filaments to printed pairs of filaments, we disentangle the effects of nozzle movement and filament extrusion. Critically, we find that capillary instabilities and interfilament fusion scale with the balance between support rheology and interfacial tension. Less viscous supports and higher interfacial tensions lead to more shrinkage and rupture at all points in the printing process, from relaxation after writing, to disturbance of the line, to writing of a second line. It is necessary to overextrude material to achieve interfilament fusion, particularly at high support viscosities and low interfacial tensions. Finally, fusion quality varies with printing orientation, and writing neighboring filaments causes displacement of existing structures. As such, specialized slicers are needed for EMB3D that consider the tighter spacings and orientation-dependent spacings necessary to achieve precise control over printed shapes.
Collapse
Affiliation(s)
- Leanne M Friedrich
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Jeremiah W Woodcock
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
30
|
Hen N, Josef E, Davidovich-Pinhas M, Levenberg S, Bianco-Peled H. On the Relation between the Viscoelastic Properties of Granular Hydrogels and Their Performance as Support Materials in Embedded Bioprinting. ACS Biomater Sci Eng 2024; 10:6734-6750. [PMID: 39344029 DOI: 10.1021/acsbiomaterials.4c01136] [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: 10/01/2024]
Abstract
Granular hydrogels, formed by jamming microgels suspension, are promising materials for three-dimensional bioprinting applications. Despite their extensive use as support materials for embedded bioprinting, the influence of the particle's physical properties on the macroscale viscoelasticity on one hand and on the printing performance on the other hand remains unclear. Herein, we investigate the linear and nonlinear rheology of κ-carrageenan granular hydrogel through small- and large-amplitude oscillatory shear measurements. We tuned the granular hydrogel's properties by changing the stiffness (soft, medium, stiff) and the packing density of the individual microgels. Characterizations in the linear viscoelasticity regime revealed that the storage modulus of granular hydrogels is not a simple function of microgel stiffness and depends on the microgel packing density. At larger strains, increasing the microgel stiffness reduced the energy dissipation of the granular beds and increased the solid-fluid transition point. To understand how the different rheological properties of granular support materials influence embedded bioprinting, we examined the printing fidelity and cellular filament shrinkage within the granular beds. We found that microgels with low packing density diminished the printing quality, while stiff microgels promoted filament roughness. In addition, we found that highly packed stiff microgels significantly reduced the postprinting contraction of cellular filaments. Overall, this work provides a comprehensive knowledge of the rheology of granular hydrogels that can be used to rationally design support beds for bioprinting applications with specific characteristics.
Collapse
Affiliation(s)
- Noy Hen
- Department of Chemical Engineering, Technion─Israel Institute of Technology, Haifa 32000, Israel
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion─Israel Institute of Technology, Haifa 32000, Israel
| | - Elinor Josef
- Technion─Israel Institute of Technology, Atlit, 12th Nahal Galim, 3033980, Israel
| | - Maya Davidovich-Pinhas
- Department of Biotechnology and Food Engineering, Technion─Israel Institute of Technology, Haifa 32000, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 32000, Israel
| | - Havazelet Bianco-Peled
- Department of Chemical Engineering, Technion─Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
31
|
Graham AJ, Khoo MW, Srivastava V, Viragova S, Kim H, Parekh K, Hennick KM, Bird M, Goldhammer N, Yu JZ, Morley CD, Lebel P, Kumar S, Rosenbluth JM, Nowakowski TJ, Klein O, Gómez-Sjöberg R, Gartner ZJ. MAGIC matrices: freeform bioprinting materials to support complex and reproducible organoid morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578324. [PMID: 38370663 PMCID: PMC10871257 DOI: 10.1101/2024.02.01.578324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Organoids are powerful models of tissue physiology, yet their applications remain limited due to their relatively simple morphology and high organoid-to-organoid structural variability. To address these limitations we developed a soft, composite yield-stress extracellular matrix that supports optimal organoid morphogenesis following freeform 3D bioprinting of cell slurries at tissue-like densities. The material is designed with two temperature regimes: at 4 °C it exhibits reversible yield-stress behavior to support long printing times without compromising cell viability. When transferred to cell culture at 37 °C, the material cross-links and exhibits similar viscoelasticity and plasticity to basement membrane extracts such as Matrigel. We first characterize the rheological properties of MAGIC matrices that optimize organoid morphogenesis, including low stiffness and high stress relaxation. Next, we combine this material with a custom piezoelectric printhead that allows more reproducible and robust self-organization from uniform and spatially organized tissue "seeds." We apply MAGIC matrix bioprinting for high-throughput generation of intestinal, mammary, vascular, salivary gland, and brain organoid arrays that are structurally similar to those grown in pure Matrigel, but exhibit dramatically improved homogeneity in organoid size, shape, maturation time, and efficiency of morphogenesis. The flexibility of this method and material enabled fabrication of fully 3D microphysiological systems, including perfusable organoid tubes that experience cyclic 3D strain in response to pressurization. Furthermore, the reproducibility of organoid structure increased the statistical power of a drug response assay by up to 8 orders-of-magnitude for a given number of comparisons. Combined, these advances lay the foundation for the efficient fabrication of complex tissue morphologies by canalizing their self-organization in both space and time.
Collapse
Affiliation(s)
- Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | | | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Sara Viragova
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
| | - Honesty Kim
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Kavita Parekh
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Kelsey M. Hennick
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Malia Bird
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Nadine Goldhammer
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jie Zeng Yu
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Cameron D. Morley
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Paul Lebel
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Sanjay Kumar
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
| | - Jennifer M. Rosenbluth
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Tomasz J. Nowakowski
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Ophir Klein
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, Los Angeles, CA
| | | | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
- Center for Cellular Construction, University of California San Francisco, San Francisco, CA
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Stankey PP, Kroll KT, Ainscough AJ, Reynolds DS, Elamine A, Fichtenkort BT, Uzel SGM, Lewis JA. Embedding Biomimetic Vascular Networks via Coaxial Sacrificial Writing into Functional Tissue. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401528. [PMID: 39092638 DOI: 10.1002/adma.202401528] [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: 01/29/2024] [Revised: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Printing human tissues and organs replete with biomimetic vascular networks is of growing interest. While it is possible to embed perfusable channels within acellular and densely cellular matrices, they do not currently possess the biomimetic architectures found in native vessels. Here, coaxial sacrificial writing into functional tissues (co-SWIFT) is developed, an embedded bioprinting method capable of generating hierarchically branching, multilayered vascular networks within both granular hydrogel and densely cellular matrices. Coaxial printheads are designed with an extended core-shell configuration to facilitate robust core-core and shell-shell interconnections between printed branching vessels during embedded bioprinting. Using optimized core-shell ink combinations, biomimetic vessels composed of a smooth muscle cell-laden shell that surrounds perfusable lumens are coaxially printed into granular matrices composed of: 1) transparent alginate microparticles, 2) sacrificial microparticle-laden collagen, or 3) cardiac spheroids derived from human induced pluripotent stem cells. Biomimetic blood vessels that exhibit good barrier function are produced by seeding these interconnected lumens with a confluent layer of endothelial cells. Importantly, it is found that co-SWIFT cardiac tissues mature under perfusion, beat synchronously, and exhibit a cardio-effective drug response in vitro. This advance opens new avenues for the scalable biomanufacturing of vascularized organ-specific tissues for drug testing, disease modeling, and therapeutic use.
Collapse
Affiliation(s)
- Paul P Stankey
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Katharina T Kroll
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexander J Ainscough
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Daniel S Reynolds
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexander Elamine
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Ben T Fichtenkort
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sebastien G M Uzel
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jennifer A Lewis
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
35
|
Jiang J, Zhu J, Lin H, Jin S, He Q, Ji W. High-Throughput Preosteoblastic Spheroids Elevate Fibroblast Growth Factor 23 via Parathyroid Hormone Signaling Pathway. Tissue Eng Part C Methods 2024; 30:402-413. [PMID: 39109940 DOI: 10.1089/ten.tec.2024.0195] [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: 08/24/2024] Open
Abstract
Fibroblast growth factor 23 (FGF23) plays a crucial role in managing renal phosphate and the synthesis of 1,25(OH)2-vitamin D3, which is essential for bone homeostasis. Developing robust in vitro systems to study FGF23-regulating mechanisms is crucial for advancing our knowledge and identifying potential therapeutic targets. The traditional in vitro 2D culture system results in relatively low expression of FGF23, complicating further exploration of its regulatory mechanisms and potential therapeutic targets. Herein, we reported a high-throughput approach to generate preosteoblastic cell spheroids with enhanced FGF23 production. For this purpose, murine preosteoblast cell line (MC3T3-E1) was cultured in our previously reported nonadherent microwells (200 µm in diameter, 148 µm in depth, and 100 µm space in between) and self-assembled into spheroids with a diameter of 92.3 ± 15.0 µm after 24 h. Compared with monolayer culture, the MC3T3-E1 spheroids showed a significant upregulation of FGF23 in both gene and protein levels after 24 h of serum-free induction. RNA sequencing and western blotting analysis further suggested that the enhanced FGF23 production in MC3T3-E1 spheroids was attributed to the activation of the parathyroid hormone (PTH)/PTH1R signaling pathway. Impressively, inhibition of PTH signaling through small molecular inhibitors or short hairpin RNA targeting PTH1R effectively reduced FGF23 production. In summary, the current study revealed the efficacy of the high-throughput formation of preosteoblast cell spheroid in stimulating FGF23 expression for mechanistic studies. Importantly, our findings highlight the potential of the current 3D spheroid system for target identification and drug discovery.
Collapse
Affiliation(s)
- Jie Jiang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jingxian Zhu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haojie Lin
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Siyu Jin
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qing He
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Figueroa-Milla AE, DeMaria W, Wells D, Jeon O, Alsberg E, Rolle MW. Vascular tissues bioprinted with smooth muscle cell-only bioinks in support baths mimic features of native coronary arteries. Biofabrication 2024; 16:045033. [PMID: 39121893 DOI: 10.1088/1758-5090/ad6d8f] [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: 03/18/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
This study explores the bioprinting of a smooth muscle cell-only bioink into ionically crosslinked oxidized methacrylated alginate (OMA) microgel baths to create self-supporting vascular tissues. The impact of OMA microgel support bath methacrylation degree and cell-only bioink dispensing parameters on tissue formation, remodeling, structure and strength was investigated. We hypothesized that reducing dispensing tip diameter from 27 G (210μm) to 30 G (159μm) for cell-only bioink dispensing would reduce tissue wall thickness and improve the consistency of tissue dimensions while maintaining cell viability. Printing with 30 G tips resulted in decreased mean wall thickness (318.6μm) without compromising mean cell viability (94.8%). Histological analysis of cell-only smooth muscle tissues cultured for 14 d in OMA support baths exhibited decreased wall thickness using 30 G dispensing tips, which correlated with increased collagen deposition and alignment. In addition, a TUNEL assay indicated a decrease in cell death in tissues printed with thinner (30 G) dispensing tips. Mechanical testing demonstrated that tissues printed with a 30 G dispensing tip exhibit an increase in ultimate tensile strength compared to those printed with a 27 G dispensing tip. Overall, these findings highlight the importance of precise control over bioprinting parameters to generate mechanically robust tissues when using cell-only bioinks dispensed and cultured within hydrogel support baths. The ability to control print dimensions using cell-only bioinks may enable bioprinting of more complex soft tissue geometries to generatein vitrotissue models.
Collapse
Affiliation(s)
- Andre E Figueroa-Milla
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| | - William DeMaria
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
| | - Derrick Wells
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Oju Jeon
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States of America
- Departments of Mechanical & Industrial Engineering, Orthopaedic Surgery, and Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, United States of America
| | - Marsha W Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States of America
- The Roux Institute at Northeastern University, Portland, ME, United States of America
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States of America
| |
Collapse
|
37
|
Alioglu MA, Yilmaz YO, Singh YP, Nagamine M, Celik N, Kim MH, Pal V, Gupta D, Ozbolat IT. Nested Biofabrication: Matryoshka-Inspired Intra-Embedded Bioprinting. SMALL METHODS 2024; 8:e2301325. [PMID: 38111377 PMCID: PMC11187694 DOI: 10.1002/smtd.202301325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/04/2023] [Indexed: 12/20/2023]
Abstract
Engineering functional tissues and organs remains a fundamental pursuit in bio-fabrication. However, the accurate constitution of complex shapes and internal anatomical features of specific organs, including their intricate blood vessels and nerves, remains a significant challenge. Inspired by the Matryoshka doll, here a new method called "Intra-Embedded Bioprinting (IEB)" is introduced building upon existing embedded bioprinting methods. a xanthan gum-based material is used which served a dual role as both a bioprintable ink and a support bath, due to its unique shear-thinning and self-healing properties. IEB's capabilities in organ modeling, creating a miniaturized replica of a pancreas using a photocrosslinkable silicone composite is demonstrated. Further, a head phantom and a Matryoshka doll are 3D printed, exemplifying IEB's capability to manufacture intricate, nested structures. Toward the use case of IEB and employing an innovative coupling strategy between extrusion-based and aspiration-assisted bioprinting, a breast tumor model that included a central channel mimicking a blood vessel, with tumor spheroids bioprinted in proximity is developed. Validation using a clinically-available chemotherapeutic drug illustrated its efficacy in reducing the tumor volume via perfusion over time. This method opens a new way of bioprinting enabling the creation of complex-shaped organs with internal anatomical features.
Collapse
Affiliation(s)
- Mecit Altan Alioglu
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Yasar Ozer Yilmaz
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- Department of Nanoscience and Nanoengineering, Istanbul Technical University, Istanbul, 34469, Turkey
| | - Yogendra Pratap Singh
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Momoka Nagamine
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Department of Chemistry, Penn State University, University Park, PA, 16802, USA
| | - Nazmiye Celik
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Myoung Hwan Kim
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Penn State University, University Park, PA, 16802, USA
| | - Vaibhav Pal
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Department of Chemistry, Penn State University, University Park, PA, 16802, USA
| | - Deepak Gupta
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Penn State University, Hershey, PA, 17033, USA
- Department of Medical Oncology, Cukurova University, Adana, 01130, Turkey
- Biotechnology Research and Application Center, Cukurova University, Adana, 01130, Turkey
| |
Collapse
|
38
|
Zhu J, He Y, Wang Y, Cai LH. Voxelated bioprinting of modular double-network bio-ink droplets. Nat Commun 2024; 15:5902. [PMID: 39003266 PMCID: PMC11246467 DOI: 10.1038/s41467-024-49705-z] [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: 09/12/2023] [Accepted: 06/17/2024] [Indexed: 07/15/2024] Open
Abstract
Analogous of pixels to two-dimensional pictures, voxels-in the form of either small cubes or spheres-are the basic building blocks of three-dimensional objects. However, precise manipulation of viscoelastic bio-ink voxels in three-dimensional space represents a grand challenge in both soft matter science and biomanufacturing. Here, we present a voxelated bioprinting technology that enables the digital assembly of interpenetrating double-network hydrogel droplets made of polyacrylamide/alginate-based or hyaluronic acid/alginate-based polymers. The hydrogels are crosslinked via additive-free and biofriendly click reaction between a pair of stoichiometrically matched polymers carrying norbornene and tetrazine groups, respectively. We develop theoretical frameworks to describe the crosslinking kinetics and stiffness of the hydrogels, and construct a diagram-of-state to delineate their mechanical properties. Multi-channel print nozzles are developed to allow on-demand mixing of highly viscoelastic bio-inks without significantly impairing cell viability. Further, we showcase the distinctive capability of voxelated bioprinting by creating highly complex three-dimensional structures such as a hollow sphere composed of interconnected yet distinguishable hydrogel particles. Finally, we validate the cytocompatibility and in vivo stability of the printed double-network scaffolds through cell encapsulation and animal transplantation.
Collapse
Affiliation(s)
- Jinchang Zhu
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Yi He
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA
| | - Yong Wang
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA
| | - Li-Heng Cai
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, VA, 22904, USA.
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22904, USA.
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
39
|
Arriola-Alvarez I, Jaunarena I, Izeta A, Lafuente H. Progenitor Cell Sources for 3D Bioprinting of Lymphatic Vessels and Potential Clinical Application. Tissue Eng Part A 2024; 30:353-366. [PMID: 37950710 DOI: 10.1089/ten.tea.2023.0204] [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: 11/13/2023] Open
Abstract
The lymphatic system maintains tissue fluid homeostasis and it is involved in the transport of nutrients and immunosurveillance. It also plays a pivotal role in both pathological and regenerative processes. Lymphatic development in the embryo occurs by polarization and proliferation of lymphatic endothelial cells from the lymph sacs, that is, lymphangiogenesis. Alternatively, lymphvasculogenesis further contributes to the formation of lymphatic vessels. In adult tissues, lymphatic formation rarely occurs under physiological conditions, being restricted to pathological processes. In lymphvasculogenesis, progenitor cells seem to be a source of lymphatic vessels. Indeed, mesenchymal stem cells, adipose stem cells, endothelial progenitor cells, and colony-forming endothelial cells are able to promote lymphatic regeneration by different mechanisms, such as direct differentiation and paracrine effects. In this review, we summarize what is known on the diverse stem/progenitor cell niches available for the lymphatic system, emphasizing the potential that these cells hold for lymphatic tissue engineering through 3D bioprinting and their translation to clinical application.
Collapse
Affiliation(s)
- Inazio Arriola-Alvarez
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| | - Ibon Jaunarena
- Gynecology Oncology Unit, Donostia University Hospital, Donostia-San Sebastián, Spain
- Obstetrics and Gynaecology Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- University of the Basque Country (UPV/EHU), Department of Medical Surgical Specialties, Leioa, Spain
| | - Ander Izeta
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- Department of Biomedical Engineering and Sciences, Tecnun-University of Navarra, Donostia-San Sebastián, Spain
| | - Héctor Lafuente
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| |
Collapse
|
40
|
Abaci A, Guvendiren M. 3D bioprinting of dense cellular structures within hydrogels with spatially controlled heterogeneity. Biofabrication 2024; 16:035027. [PMID: 38821144 DOI: 10.1088/1758-5090/ad52f1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/31/2024] [Indexed: 06/02/2024]
Abstract
Embedded bioprinting is an emerging technology for precise deposition of cell-laden or cell-only bioinks to construct tissue like structures. Bioink is extruded or transferred into a yield stress hydrogel or a microgel support bath allowing print needle motion during printing and providing temporal support for the printed construct. Although this technology has enabled creation of complex tissue structures, it remains a challenge to develop a support bath with user-defined extracellular mimetic cues and their spatial and temporal control. This is crucial to mimic the dynamic nature of the native tissue to better regenerate tissues and organs. To address this, we present a bioprinting approach involving printing of a photocurable viscous support layer and bioprinting of a cell-only or cell-laden bioink within this viscous layer followed by brief exposure to light to partially crosslink the support layer. This approach does not require shear thinning behavior and is suitable for a wide range of photocurable hydrogels to be used as a support. It enables multi-material printing to spatially control support hydrogel heterogeneity including temporal delivery of bioactive cues (e.g. growth factors), and precise patterning of dense multi-cellular structures within these hydrogel supports. Here, dense stem cell aggregates are printed within methacrylated hyaluronic acid-based hydrogels with patterned heterogeneity to spatially modulate human mesenchymal stem cell osteogenesis. This study has significant impactions on creating tissue interfaces (e.g. osteochondral tissue) in which spatial control of extracellular matrix properties for patterned stem cell differentiation is crucial.
Collapse
Affiliation(s)
- Alperen Abaci
- Otto H. York Chemical and Materials Engineering Department, New Jersey Institute of Technology, Newark, NJ, United States of America
| | - Murat Guvendiren
- Otto H. York Chemical and Materials Engineering Department, New Jersey Institute of Technology, Newark, NJ, United States of America
- Bioengineering Department, New Jersey Institute of Technology, Newark, NJ, United States of America
| |
Collapse
|
41
|
Karyappa R, Nagaraju N, Yamagishi K, Koh XQ, Zhu Q, Hashimoto M. 3D printing of polyvinyl alcohol hydrogels enabled by aqueous two-phase system. MATERIALS HORIZONS 2024; 11:2701-2717. [PMID: 38506347 DOI: 10.1039/d3mh01714a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
The synthesis of PVA hydrogels (PVA-Hy) requires a highly basic environment (e.g., an aqueous solution of sodium hydroxide, NaOH, 14% w/w, 4.2 M), but the rapid crosslinking of PVA due to high pH makes it challenging to perform layer-by-layer three-dimensional (3D) printing of PVA-Hy. This work demonstrated 3D printing of PVA-Hy in moderate alkaline conditions (e.g., NaOH, 1% w/w, 0.3 M) assisted by aqueous two-phase system (ATPS). Salting out of PVA to form ATPS allowed temporal shape retention of a 3D-printed PVA structure while it was physically crosslinked in moderate alkaline conditions. Crucially, the layer-to-layer adhesion of PVA was facilitated by delayed crosslinking of PVA that required additional reaction time and overlapping between the layers. To verify this principle, we studied the feasibility of direct ink write (DIW) 3D printing of PVA inks (5-25% w/w, μ = 0.1-20 Pa s, and MW = 22 000 and 74 800) in aqueous embedding media offering three distinct chemical environments: (1) salts for salting out (e.g., Na2SO4), (2) alkali hydroxides for physical crosslinking (e.g., NaOH), and (3) a mixture of salt and alkali hydroxide. Our study suggested the feasibility of 3D-printed PVA-Hy using the mixture of salt and alkali hydroxide, demonstrating a unique concept of embedded 3D printing enabled by ATPS for temporary stabilization of the printed structures to facilitate 3D fabrication.
Collapse
Affiliation(s)
- Rahul Karyappa
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8, Somapah Road, Singapore 487372, Republic of Singapore.
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Republic of Singapore
| | - Nidhi Nagaraju
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8, Somapah Road, Singapore 487372, Republic of Singapore.
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8, Somapah Road, Singapore 487372, Republic of Singapore
| | - Kento Yamagishi
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8, Somapah Road, Singapore 487372, Republic of Singapore.
| | - Xue Qi Koh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Republic of Singapore
| | - Qiang Zhu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Republic of Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Michinao Hashimoto
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8, Somapah Road, Singapore 487372, Republic of Singapore.
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8, Somapah Road, Singapore 487372, Republic of Singapore
| |
Collapse
|
42
|
Sreepadmanabh M, Arun AB, Bhattacharjee T. Design approaches for 3D cell culture and 3D bioprinting platforms. BIOPHYSICS REVIEWS 2024; 5:021304. [PMID: 38765221 PMCID: PMC11101206 DOI: 10.1063/5.0188268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/01/2024] [Indexed: 05/21/2024]
Abstract
The natural habitat of most cells consists of complex and disordered 3D microenvironments with spatiotemporally dynamic material properties. However, prevalent methods of in vitro culture study cells under poorly biomimetic 2D confinement or homogeneous conditions that often neglect critical topographical cues and mechanical stimuli. It has also become increasingly apparent that cells in a 3D conformation exhibit dramatically altered morphological and phenotypical states. In response, efforts toward designing biomaterial platforms for 3D cell culture have taken centerstage over the past few decades. Herein, we present a broad overview of biomaterials for 3D cell culture and 3D bioprinting, spanning both monolithic and granular systems. We first critically evaluate conventional monolithic hydrogel networks, with an emphasis on specific experimental requirements. Building on this, we document the recent emergence of microgel-based 3D growth media as a promising biomaterial platform enabling interrogation of cells within porous and granular scaffolds. We also explore how jammed microgel systems have been leveraged to spatially design and manipulate cellular structures using 3D bioprinting. The advent of these techniques heralds an unprecedented ability to experimentally model complex physiological niches, with important implications for tissue bioengineering and biomedical applications.
Collapse
Affiliation(s)
- M Sreepadmanabh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| | - Ashitha B. Arun
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| | - Tapomoy Bhattacharjee
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| |
Collapse
|
43
|
Navara AM, Xu Y, Perez MR, Mikos AG. Aspects of a Suspended Bioprinting System Affect Cell Viability and Support Bath Properties. Tissue Eng Part A 2024; 30:256-269. [PMID: 37341034 DOI: 10.1089/ten.tea.2023.0097] [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: 06/22/2023] Open
Abstract
Suspended hydrogel printing is a growing method for fabricating bioprinted hydrogel constructs, largely due to how it enables nonviscous hydrogel inks to be used in extrusion printing. In this work, a previously developed poly(N-isopropylacrylamide)-based thermogelling suspended bioprinting system was examined in the context of chondrocyte-laden printing. Material factors such as ink concentration and cell concentration were found to have a significant effect on printed chondrocyte viability. In addition, the heated poloxamer support bath was able to maintain chondrocyte viability for up to 6 h of residence within the bath. The relationship between the ink and support bath was also assessed by measuring the rheological properties of the bath before and after printing. Bath storage modulus and yield stress decreased during printing as nozzle size was reduced, indicating the likelihood that dilution occurs over time through osmotic exchange with the ink. Altogether this work demonstrates the promise for printing high-resolution cell-encapsulating tissue engineering constructs, while also elucidating complex relationships between the ink and bath, which must be taken into consideration when designing suspended printing systems.
Collapse
Affiliation(s)
- Adam M Navara
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Yilan Xu
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Marissa R Perez
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas, USA
| |
Collapse
|
44
|
Wu Y, Yang X, Gupta D, Alioglu MA, Qin M, Ozbolat V, Li Y, Ozbolat IT. Dissecting the Interplay Mechanism among Process Parameters toward the Biofabrication of High-Quality Shapes in Embedded Bioprinting. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2313088. [PMID: 38952568 PMCID: PMC11216718 DOI: 10.1002/adfm.202313088] [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: 10/23/2023] [Indexed: 07/03/2024]
Abstract
Embedded bioprinting overcomes the barriers associated with the conventional extrusion-based bioprinting process as it enables the direct deposition of bioinks in 3D inside a support bath by providing in situ self-support for deposited bioinks during bioprinting to prevent their collapse and deformation. Embedded bioprinting improves the shape quality of bioprinted constructs made up of soft materials and low-viscosity bioinks, leading to a promising strategy for better anatomical mimicry of tissues or organs. Herein, the interplay mechanism among the printing process parameters toward improved shape quality is critically reviewed. The impact of material properties of the support bath and bioink, printing conditions, cross-linking mechanisms, and post-printing treatment methods, on the printing fidelity, stability, and resolution of the structures is meticulously dissected and thoroughly discussed. Further, the potential scope and applications of this technology in the fields of bioprinting and regenerative medicine are presented. Finally, outstanding challenges and opportunities of embedded bioprinting as well as its promise for fabricating functional solid organs in the future are discussed.
Collapse
Affiliation(s)
- Yang Wu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China
| | - Xue Yang
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China
| | - Deepak Gupta
- The Huck Institutes of the Life Sciences, Penn State University University Park, PA 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
| | - Mecit Altan Alioglu
- The Huck Institutes of the Life Sciences, Penn State University University Park, PA 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
| | - Minghao Qin
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China
| | - Veli Ozbolat
- Biotechnology Research and Application Center, Cukurova University, Adana 01130, Turkey
- Ceyhan Engineering Faculty, Mechanical Engineering Department, Cukurova University, Adana 01330, Turkey
- Institute of Natural and Applied Sciences, Tissue Engineering Department, Cukurova University, Adana 01130, Turkey
| | - Yao Li
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University University Park, PA 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, Penn State University, University Park, PA 16802, USA
- Materials Research Institute, Penn State University, University Park, PA 16802, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Penn State University, Hershey, PA 17033, USA
| |
Collapse
|
45
|
Lai G, Meagher L. Versatile xanthan gum-based support bath material compatible with multiple crosslinking mechanisms: rheological properties, printability, and cytocompatibility study. Biofabrication 2024; 16:035005. [PMID: 38565131 DOI: 10.1088/1758-5090/ad39a8] [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: 10/05/2023] [Accepted: 04/02/2024] [Indexed: 04/04/2024]
Abstract
Extrusion-based bioprinting is a promising technology for the fabrication of complex three-dimensional (3D) tissue-engineered constructs. To further improve the printing accuracy and provide mechanical support during the printing process, hydrogel-based support bath materials have been developed. However, the gel structure of some support bath materials can be compromised when exposed to certain bioink crosslinking cues, hence their compatibility with bioinks can be limited. In this study, a xanthan gum-based composite support material compatible with multiple crosslinking mechanisms is developed. Different support bath materials can have different underlying polymeric structures, for example, particulate suspensions and polymer solution with varying supramolecular structure) and these properties are governed by a variety of different intermolecular interactions. However, common rheological behavior can be expected because they have similar demonstrated performance and functionality. To provide a detailed exploration/identification of the common rheological properties expressed by different support bath materials from a unified perspective, benchmark support bath materials from previous studies were prepared. A comparative rheological study revealed both the structural and shear behavior characteristics shared by support bath materials, including yield stress, gel complex moduli, shear-thinning behavior, and self-healing properties. Gel structural stability and functionality of support materials were tested in the presence of various crosslinking stimuli, confirming the versatility of the xanthan-based support material. We further investigated the effect of support materials and the diameter of extrusion needles on the printability of bioinks to demonstrate the improvement in bioink printability and structural integrity. Cytotoxicity and cell encapsulation viability tests were carried out to confirm the cell compatibility of the xanthan gum-based support bath material. We propose and demonstrate the versatility and compatibility of the novel support bath material and provide detailed new insight into the essential properties and behavior of these materials that serve as a guide for further development of support bath-based 3D bioprinting.
Collapse
Affiliation(s)
- Guanyu Lai
- Department of Materials Science and Engineering, Monash University, Clayton, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Australia
| |
Collapse
|
46
|
Du J, Lee S, Sinha S, Solberg FS, Ho DLL, Sampson JP, Wang Q, Tam T, Skylar-Scott MA. A Visual, In-Expensive, and Wireless Capillary Rheometer for Characterizing Wholly-Cellular Bioinks. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304778. [PMID: 38085139 PMCID: PMC11545891 DOI: 10.1002/smll.202304778] [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: 06/06/2023] [Revised: 10/14/2023] [Indexed: 08/15/2024]
Abstract
Rheological measurements with in situ visualization can elucidate the microstructural origin of complex flow behaviors of an ink. However, existing commercial rheometers suffer from high costs, the need for dedicated facilities for microfabrication, a lack of design flexibility, and cabling that complicates operation in sterile or enclosed environments. To address these limitations, a low-cost ($300) visual, in-expensive and wireless rheometer (VIEWR) using 3D-printed and off-the-shelf components is presented. VIEWR measurements are validated by steady-state and transient flow responses for different complex fluids, and microstructural flow profiles and evolution of yield-planes are revealed via particle image velocimetry. Using the VIEWR, a wholly-cellular bioink system comprised of compacted cell aggregates is characterized, and complex yield-stress and viscoelastic responses are captured via concomitantly visualizing the spatiotemporal evolution of aggregate morphology. A symmetric hyperbolic extensional-flow geometry is further constructed inside a capillary tube using digital light processing. Such geometries allow for measuring the extensional viscosity at varying deformation rates and further visualizing the alignment and stretching of aggregates under external flow. Synchronized but asymmetric evolution of aggregate orientation and strain through the neck is visualized. Using varying geometries, the jamming and viscoelastic deformation of aggregates are shown to contribute to the extensional viscosity of the wholly-cellular bioinks.
Collapse
Affiliation(s)
- Jianyi Du
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Stacey Lee
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Soham Sinha
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Fredrik S Solberg
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Debbie L L Ho
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Joshua P Sampson
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Qiuling Wang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Tony Tam
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Mark A Skylar-Scott
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Basic Science and Engineering Initiative, Children's Heart Center, Stanford University, Stanford, CA, 94304, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| |
Collapse
|
47
|
Zhang C, Hua W, Mitchell K, Raymond L, Delzendehrooy F, Wen L, Do C, Chen J, Yang Y, Linke G, Zhang Z, Krishnan MA, Kuss M, Coulter R, Bandala E, Liao Y, Duan B, Zhao D, Chai G, Jin Y. Multiscale embedded printing of engineered human tissue and organ equivalents. Proc Natl Acad Sci U S A 2024; 121:e2313464121. [PMID: 38346211 PMCID: PMC10907305 DOI: 10.1073/pnas.2313464121] [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: 08/10/2023] [Accepted: 01/09/2024] [Indexed: 02/15/2024] Open
Abstract
Creating tissue and organ equivalents with intricate architectures and multiscale functional feature sizes is the first step toward the reconstruction of transplantable human tissues and organs. Existing embedded ink writing approaches are limited by achievable feature sizes ranging from hundreds of microns to tens of millimeters, which hinders their ability to accurately duplicate structures found in various human tissues and organs. In this study, a multiscale embedded printing (MSEP) strategy is developed, in which a stimuli-responsive yield-stress fluid is applied to facilitate the printing process. A dynamic layer height control method is developed to print the cornea with a smooth surface on the order of microns, which can effectively overcome the layered morphology in conventional extrusion-based three-dimensional bioprinting methods. Since the support bath is sensitive to temperature change, it can be easily removed after printing by tuning the ambient temperature, which facilitates the fabrication of human eyeballs with optic nerves and aortic heart valves with overhanging leaflets on the order of a few millimeters. The thermosensitivity of the support bath also enables the reconstruction of the full-scale human heart on the order of tens of centimeters by on-demand adding support bath materials during printing. The proposed MSEP demonstrates broader printable functional feature sizes ranging from microns to centimeters, providing a viable and reliable technical solution for tissue and organ printing in the future.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
- State Key Laboratory of High-Performance Precision Manufacturing, School of Mechanical Engineering, Dalian University of Technology, Dalian116024, China
| | - Weijian Hua
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
| | - Kellen Mitchell
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
| | - Lily Raymond
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
| | - Fatemeh Delzendehrooy
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, IA50011
| | - Lai Wen
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, School of Medicine, University of Nevada, Reno, NV89557
| | - Changwoo Do
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN37831-6475
| | - Jihua Chen
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN37830
| | - Ying Yang
- Department of Chemistry, University of Nevada, Reno, NV89557
| | - Gabe Linke
- Three-Dimensional Advanced Visualization Laboratory, Department of Pediatric Radiology, Children’s Hospital & Medical Center, Omaha, NE68114
| | - Zhengyi Zhang
- School of Naval Architecture and Ocean Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Mena Asha Krishnan
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE68198
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE68198
| | - Ryan Coulter
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
| | - Erick Bandala
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
| | - Yiliang Liao
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, IA50011
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE68198
| | - Danyang Zhao
- State Key Laboratory of High-Performance Precision Manufacturing, School of Mechanical Engineering, Dalian University of Technology, Dalian116024, China
| | - Guangrui Chai
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang110004, China
| | - Yifei Jin
- Department of Mechanical Engineering, University of Nevada, Reno, NV89557
| |
Collapse
|
48
|
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
|
49
|
Zhang H, Luo Y, Hu Z, Chen M, Chen S, Yao Y, Yao J, Shao X, Wu K, Zhu Y, Fu J. Cation-crosslinked κ-carrageenan sub-microgel medium for high-quality embedded bioprinting. Biofabrication 2024; 16:025009. [PMID: 38198708 DOI: 10.1088/1758-5090/ad1cf3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Three-dimensional (3D) bioprinting embedded within a microgel bath has emerged as a promising strategy for creating intricate biomimetic scaffolds. However, it remains a great challenge to construct tissue-scale structures with high resolution by using embedded 3D bioprinting due to the large particle size and polydispersity of the microgel medium, as well as its limited cytocompatibility. To address these issues, novel uniform sub-microgels of cell-friendly cationic-crosslinked kappa-carrageenan (κ-Car) are developed through an easy-to-operate mechanical grinding strategy. Theseκ-Car sub-microgels maintain a uniform submicron size of around 642 nm and display a rapid jamming-unjamming transition within 5 s, along with excellent shear-thinning and self-healing properties, which are critical for the high resolution and fidelity in the construction of tissue architecture via embedded 3D bioprinting. Utilizing this new sub-microgel medium, various intricate 3D tissue and organ structures, including the heart, lungs, trachea, branched vasculature, kidney, auricle, nose, and liver, are successfully fabricated with delicate fine structures and high shape fidelity. Moreover, the bone marrow mesenchymal stem cells encapsulated within the printed constructs exhibit remarkable viability exceeding 92.1% and robust growth. Thisκ-Car sub-microgel medium offers an innovative avenue for achieving high-quality embedded bioprinting, facilitating the fabrication of functional biological constructs with biomimetic structural organizations.
Collapse
Affiliation(s)
- Hua Zhang
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, Zhejiang 310027, People's Republic of China
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
| | - Yang Luo
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
| | - Zeming Hu
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Mengxi Chen
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Shang Chen
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Yudong Yao
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Jie Yao
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
| | - Xiaoqi Shao
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
| | - Kerong Wu
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, People's Republic of China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Jun Fu
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510275, People's Republic of China
| |
Collapse
|
50
|
Budharaju H, Sundaramurthi D, Sethuraman S. Embedded 3D bioprinting - An emerging strategy to fabricate biomimetic & large vascularized tissue constructs. Bioact Mater 2024; 32:356-384. [PMID: 37920828 PMCID: PMC10618244 DOI: 10.1016/j.bioactmat.2023.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/16/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Three-dimensional bioprinting is an advanced tissue fabrication technique that allows printing complex structures with precise positioning of multiple cell types layer-by-layer. Compared to other bioprinting methods, extrusion bioprinting has several advantages to print large-sized tissue constructs and complex organ models due to large build volume. Extrusion bioprinting using sacrificial, support and embedded strategies have been successfully employed to facilitate printing of complex and hollow structures. Embedded bioprinting is a gel-in-gel approach developed to overcome the gravitational and overhanging limits of bioprinting to print large-sized constructs with a micron-scale resolution. In embedded bioprinting, deposition of bioinks into the microgel or granular support bath will be facilitated by the sol-gel transition of the support bath through needle movement inside the granular medium. This review outlines various embedded bioprinting strategies and the polymers used in the embedded systems with advantages, limitations, and efficacy in the fabrication of complex vascularized tissues or organ models with micron-scale resolution. Further, the essential requirements of support bath systems like viscoelasticity, stability, transparency and easy extraction to print human scale organs are discussed. Additionally, the organs or complex geometries like vascular constructs, heart, bone, octopus and jellyfish models printed using support bath assisted printing methods with their anatomical features are elaborated. Finally, the challenges in clinical translation and the future scope of these embedded bioprinting models to replace the native organs are envisaged.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Center for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Center, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|