1
|
Islam MA, Hasan MN, Evan MSH, Uddin MJ, Tulin WS, Islam MS, Khandaker MU, Rahman IMM, Chowdhury FI. Chitin nanofibers: recent advances in preparation and applications in biomedical and beyond. RSC Adv 2025; 15:14655-14690. [PMID: 40390794 PMCID: PMC12086821 DOI: 10.1039/d4ra06937d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/01/2025] [Indexed: 05/21/2025] Open
Abstract
Chitin and chitosan-based nanofibers (ChNFs), derived from renewable sources, have emerged as promising biomaterials due to their unique properties such as high surface area, porosity, biocompatibility, and biodegradability. This review provides a comprehensive overview of ChNF extraction and synthesis, focusing on both top-down and bottom-up approaches. A comparative analysis of these methods is presented, highlighting the challenges, opportunities, environmental impact, cost-effectiveness, and quality consistency associated with each. The advantages of ChNFs over similar nanomaterials are elucidated, emphasizing their diverse applications in biomedical and environmental fields. Biomedical applications include drug delivery, tissue engineering, cancer treatment, wound healing, and biosensing. Environmental applications encompass water treatment, air filtration, agriculture, and biodegradable packaging. Despite their potential, challenges remain, including low solubility, unstable mechanical properties, and inconsistent quality, which limit their widespread use. This review also examines recent advancements in ChNF research, aiming to guide the development of efficient and environmentally friendly synthesis methods. By encouraging innovation in ChNF-based nanotechnologies, this research contributes to a more sustainable future.
Collapse
Affiliation(s)
- M Ariful Islam
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
- Graduate School of Natural Science and Technology, Kanazawa University Kakuma Kanazawa 920-1192 Japan
| | - M Nahid Hasan
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
| | - M Sadik Hussain Evan
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
| | - M Jalal Uddin
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
| | - Wahid Salekin Tulin
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
| | - M Saydul Islam
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
| | - Mayeen Uddin Khandaker
- Applied Physics and Radiation Technologies Group, CCDCU, Faculty of Engineering and Technology, Sunway University Bandar Sunway 47500 Selangor Malaysia
- Department of Physics, College of Science, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Republic of Korea
- Faculty of Graduate Studies, Daffodil International University Daffodil Smart City, Birulia, Savar Dhaka 1216 Bangladesh
| | - Ismail M M Rahman
- Institute of Environmental Radioactivity, Fukushima University 1 Kanayagawa Fukushima City Fukushima 960-1296 Japan
| | - Faisal I Chowdhury
- Nanotechnology, Renewable Energy and Catalysis Laboratory, Department of Chemistry, University of Chittagong Chattogram 4331 Bangladesh
| |
Collapse
|
2
|
Abou-Shanab AM, Gaser OA, Soliman MW, Oraby A, Salah RA, Gabr M, Edris AAF, Mohamed I, El-Badri N. Human amniotic membrane scaffold enhances adipose mesenchymal stromal cell mitochondrial bioenergetics promoting their regenerative capacities. Mol Cell Biochem 2025; 480:2611-2632. [PMID: 39453499 DOI: 10.1007/s11010-024-05094-x] [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: 04/15/2024] [Accepted: 08/12/2024] [Indexed: 10/26/2024]
Abstract
The human amniotic membrane (hAM) has been applied as a scaffold in tissue engineering to sustain stem cells and enhance their regenerative capacities. We investigated the molecular and biochemical regulations of mesenchymal stromal cells (MSCs) cultured on hAM scaffold in a three-dimensional (3D) setting. Culture of adipose-MSCs (AMSCs) on decellularized hAM showed significant improvement in their viability, proliferative capacity, resistance to apoptosis, and enhanced MSC markers expression. These cultured MSCs displayed altered expression of markers associated with pro-angiogenesis and inflammation and demonstrated increased potential for differentiation into adipogenic and osteogenic lineages. The hAM scaffold modulated cellular respiration by upregulating glycolysis in MSCs as evidenced by increased glucose consumption, cellular pyruvate and lactate production, and upregulation of glycolysis markers. These metabolic changes modulated mitochondrial oxidative phosphorylation (OXPHOS) and altered the production of reactive oxygen species (ROS), expression of OXPHOS markers, and total antioxidant capacity. They also significantly boosted the urea cycle and altered the mitochondrial ultrastructure. Similar findings were observed in bone marrow-derived MSCs (BMSCs). Live cell imaging of BMSCs cultured in the same 3D environment revealed dynamic changes in cellular activity and interactions with its niche. These findings provide evidence for the favorable properties of hAM as a biomimetic scaffold for enhancing the in vitro functionality of MSCs and supporting their potential usefulness in clinical applications.
Collapse
Affiliation(s)
- Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ola A Gaser
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Mariam Waleed Soliman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Alaa Oraby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | | | - Ihab Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt.
| |
Collapse
|
3
|
Petit N, Chang YYJ, Lobianco FA, Hodgkinson T, Browne S. Hyaluronic acid as a versatile building block for the development of biofunctional hydrogels: In vitro models and preclinical innovations. Mater Today Bio 2025; 31:101596. [PMID: 40083836 PMCID: PMC11903855 DOI: 10.1016/j.mtbio.2025.101596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Hyaluronic acid (HyA) is a non-sulphated linear polysaccharide found abundantly in the extracellular matrix, known for its biocompatibility and versatility in tissue engineering. Chemical modifications of HyA, including methacrylate, acrylate, click chemistry, norbornene, or host-guest chemistry, are necessary for the formation of stable hydrogels with tuneable biophysical characteristics. These modifications enable precise control over stiffness, swelling, degradation, and advanced functionalities such as shear-thinning, self-healing, and injectability. Functionalisation further enhances hydrogel bioactivity, enabling controlled cell adhesion, modulation of cell behaviour, hydrogel degradation, and release profiles, as well as inflammation modulation or bacterial growth inhibition. These are achieved by conjugating proteins, peptides, antibodies, or reactive chemical groups. HyA hydrogels find broad applications both in vitro and in vivo. In vitro, HyA-based hydrogels can support the development of models to understand fundamental processes in health and mechanisms behind disease progression, serving as highly tuneable extracellular matrix mimetics. As therapeutic interventions, injectable or implantable HyA-based hydrogels have been developed to repair a range of tissues, including cartilage, bone, muscle, and skin defects. However, issues remain to be addressed before widespread adoption of HyA-based hydrogels as clinical options. Future innovations for HyA hydrogels include its establishment as an enabling technology for the delivery of novel therapeutics, with a particular focus on immunomodulatory molecules, and the development of more dynamic, tissue-mimetic HyA-based hydrogels.
Collapse
Affiliation(s)
- Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Yu-yin Joanne Chang
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Franz Acker Lobianco
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Tom Hodgkinson
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
4
|
Mei X, Yang Z, Wang X, Shi A, Blanchard J, Elahi F, Kang H, Orive G, Zhang YS. Integrating microfluidic and bioprinting technologies: advanced strategies for tissue vascularization. LAB ON A CHIP 2025; 25:764-786. [PMID: 39775452 DOI: 10.1039/d4lc00280f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Tissue engineering offers immense potential for addressing the unmet needs in repairing tissue damage and organ failure. Vascularization, the development of intricate blood vessel networks, is crucial for the survival and functions of engineered tissues. Nevertheless, the persistent challenge of ensuring an ample nutrient supply within implanted tissues remains, primarily due to the inadequate formation of blood vessels. This issue underscores the vital role of the human vascular system in sustaining cellular functions, facilitating nutrient exchange, and removing metabolic waste products. In response to this challenge, new approaches have been explored. Microfluidic devices, emulating natural blood vessels, serve as valuable tools for investigating angiogenesis and allowing the formation of microvascular networks. In parallel, bioprinting technologies enable precise placement of cells and biomaterials, culminating in vascular structures that closely resemble the native vessels. To this end, the synergy of microfluidics and bioprinting has further opened up exciting possibilities in vascularization, encompassing innovations such as microfluidic bioprinting. These advancements hold great promise in regenerative medicine, facilitating the creation of functional tissues for applications ranging from transplantation to disease modeling and drug testing. This review explores the potentially transformative impact of microfluidic and bioprinting technologies on vascularization strategies within the scope of tissue engineering.
Collapse
Affiliation(s)
- Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| | - Ziyi Yang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
- School of Biological Science, University of California Irvine, Irvine, CA 92697, USA
| | - Xiran Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA 92161, USA
| | - Alan Shi
- Brookline High School, Brookline, MA 02445, USA
| | - Joel Blanchard
- Departments of Neurology, Neuroscience, and Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fanny Elahi
- Departments of Neurology, Neuroscience, and Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea.
- College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, 01007, Spain
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Yin Q, Song H, Wang Z, Ma Z, Zhang W. Acoustic black hole effect enhanced micro-manipulator. MICROSYSTEMS & NANOENGINEERING 2024; 10:144. [PMID: 39394206 PMCID: PMC11470035 DOI: 10.1038/s41378-024-00789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/25/2024] [Accepted: 08/22/2024] [Indexed: 10/13/2024]
Abstract
Microparticle manipulation is a critical concern across various fields including microfabrication, flexible electronics and tissue engineering. Acoustic-activated sharp structures have been designed as simple and flexible tools to manipulate microparticles with their good compatibility, fast response, and broad tunability. However, there still lacks rational acoustic-structure design for effective energy concentration at the acoustic-activated sharp structures for microparticle manipulation. Here, we present the acoustic black hole (ABH) effect as enhancement for the acoustic micro-manipulator. It provides great reliability, simplicity and ease of use, supporting custom design of high-throughput patterning modes. Moreover, compared to commonly used configurations, such as cylindrical or conical microneedles, those microneedles with ABH profile exhibit superior acoustic energy focusing at the tip and induce stronger acoustofluidic effects. The average acoustic flow velocity induced by the ABH microneedle is 154 times greater than that of the conical one and 45 times greater than that of the cylindrical microneedle. Besides, the average acoustic radiation force (ARF) produced by the ABH microneedle against acrylic microparticles is about 319 times greater than that of the cylindrical one and 16 times greater than that of the conical one. These results indicate that ABH design significantly enhances microparticle manipulation. We demonstrate this concept with ABH effect enhanced microparticle manipulation and study the parameters influencing its performance including operating frequency, operating voltage and particle diameter. Furthermore, considering the flexibility of this system, we employ it for various patterning and high-throughput microparticle manipulation. This work paves the way for controllable microparticle manipulation, holding great potential for applications in microfabrication and biomedicine.
Collapse
Affiliation(s)
- Qiu Yin
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyong Song
- College of Mechanical and Vehicle Engineering, Hunan University, Changsha, China
| | - Zhaolong Wang
- School of Energy Science and Engineering, Harbin Institute of Technology, Harbin, P. R. China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Wenming Zhang
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
8
|
Bathrinarayanan PV, Hallam SM, Grover LM, Vigolo D, Simmons MJH. Microfluidics as a Powerful Tool to Investigate Microvascular Dysfunction in Trauma Conditions: A Review of the State-of-the-Art. Adv Biol (Weinh) 2024; 8:e2400037. [PMID: 39031943 DOI: 10.1002/adbi.202400037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/18/2024] [Indexed: 07/22/2024]
Abstract
Skeletal muscle trauma such as fracture or crush injury can result in a life-threatening condition called acute compartment syndrome (ACS), which involves elevated compartmental pressure within a closed osteo-fascial compartment, leading to collapse of the microvasculature and resulting in necrosis of the tissue due to ischemia. Diagnosis of ACS is complex and controversial due to the lack of standardized objective methods, which results in high rates of misdiagnosis/late diagnosis, leading to permanent neuro-muscular damage. ACS pathophysiology is poorly understood at a cellular level due to the lack of physiologically relevant models. In this context, microfluidics organ-on-chip systems (OOCs) provide an exciting opportunity to investigate the cellular mechanisms of microvascular dysfunction that leads to ACS. In this article, the state-of-the-art OOCs designs and strategies used to investigate microvasculature dysfunction mechanisms is reviewed. The differential effects of hemodynamic shear stress on endothelial cell characteristics such as morphology, permeability, and inflammation, all of which are altered during microvascular dysfunction is highlighted. The article then critically reviews the importance of microfluidics to investigate closely related microvascular pathologies that cause ACS. The article concludes by discussing potential biomarkers of ACS with a special emphasis on glycocalyx and providing a future perspective.
Collapse
Affiliation(s)
- P Vasanthi Bathrinarayanan
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - S M Hallam
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
| | - L M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - D Vigolo
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - M J H Simmons
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
| |
Collapse
|
9
|
Tang T, Zhang P, Zhang Q, Man X, Xu Y. Fabrication of heterocellular spheroids with controllable core-shell structure using inertial focusing effect for scaffold-free 3D cell culture models. Biofabrication 2024; 16:045013. [PMID: 39019062 DOI: 10.1088/1758-5090/ad647e] [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: 03/31/2024] [Accepted: 07/17/2024] [Indexed: 07/19/2024]
Abstract
Three-dimensional (3D) cell culture models capable of emulating the biological functions of natural tissues are pivotal in tissue engineering and regenerative medicine. Despite progress, the fabrication ofin vitroheterocellular models that mimic the intricate structures of natural tissues remains a significant challenge. In this study, we introduce a novel, scaffold-free approach leveraging the inertial focusing effect in rotating hanging droplets for the reliable production of heterocellular spheroids with controllable core-shell structures. Our method offers precise control over the core-shell spheroid's size and geometry by adjusting the cell suspension density and droplet morphology. We successfully applied this technique to create hair follicle organoids, integrating dermal papilla cells within the core and epidermal cells in the shell, thereby achieving markedly enhanced hair inducibility compared to mixed-structure models. Furthermore, we have developed melanoma tumor spheroids that accurately mimic the dynamic interactions between tumor and stromal cells, showing increased invasion capabilities and altered expressions of cellular adhesion molecules and proteolytic enzymes. These findings underscore the critical role of cellular spatial organization in replicating tissue functionalityin vitro. Our method represents a significant advancement towards generating heterocellular spheroids with well-defined architectures, offering broad implications for biological research and applications in tissue engineering.
Collapse
Affiliation(s)
- Tan Tang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, People's Republic of China
| | - Pengfei Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, People's Republic of China
| | - Qiuting Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, People's Republic of China
| | - Xingkun Man
- School of Physics, Beihang University, Beijing, People's Republic of China
| | - Ye Xu
- School of Mechanical Engineering and Automation, Beihang University, Beijing, People's Republic of China
| |
Collapse
|
10
|
Fardous RS, Alshmmari S, Tawfik E, Khadra I, Ramadan Q, Zourob M. An Integrated and Modular Compartmentalized Microfluidic System with Tunable Electrospun Porous Membranes for Epithelialized Organs-on-a-Chip. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39047263 DOI: 10.1021/acsami.4c08864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
A modular and 3D compartmentalized microfluidic system with electrospun porous membranes (PMs) for epithelialized organ-on-a-chip systems is presented. Our novel approach involves direct deposition of polymer nanofibers onto a patterned poly(methyl methacrylate) (PMMA) substrate using electrospinning, resulting in an integrated PM within the microfluidic chip. The in situ deposition of the PM eliminates the need for additional assembly processes. To demonstrate the high throughput membrane integration capability of our approach, we successfully deposited nanofibers onto various chip designs with complex microfluidic planar structures and expanded dimensions. We characterized and tested the fully PMMA chip by growing an epithelial monolayer using the Caco-2 cell line to study drug permeability. A comprehensive analysis of the bulk and surface properties of the membrane's fibers made of PMMA and polystyrene (PS) was conducted to determine the polymer with the best performance for cell culture and drug transport applications. The PMMA-based membrane, with a PMMA/PVP ratio of 5:1, allowed for the fabrication of a uniform membrane structure along the aligned nanofibers. By modulating the fiber diameter and total thickness of the membrane, we could adjust the membrane's porosity for specific cell culture applications. The PMMA-PVP nanofibers exhibited a low polydispersity index value, indicating monodispersed nanofibers and a more homogeneous and uniform fiber network. Both types of membranes demonstrated excellent mechanical integrity under medium perfusion flow rates. However, the PMMA-PVP composition offered a tailored porous structure with modulable porosity based on the fiber diameter and thickness. Our developed platform enables dynamic in vitro modeling of the epithelial barrier and has applications in drug transport and in vitro microphysiological systems.
Collapse
Affiliation(s)
- Roa S Fardous
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow G4 0RE, U.K
- Alfaisal University, Riyadh 11533, Kingdome Saudi Arabia
| | - Sultan Alshmmari
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, Jena 07745, Germany
- Alfaisal University, Riyadh 11533, Kingdome Saudi Arabia
| | - Essam Tawfik
- Advanced Diagnostics & Therapeutics Institute, King Abdulaziz City for Science and Technology, Riyadh 12354, Kingdome Saudi Arabia
| | - Ibrahim Khadra
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow G4 0RE, U.K
| | - Qasem Ramadan
- Alfaisal University, Riyadh 11533, Kingdome Saudi Arabia
| | | |
Collapse
|
11
|
Zhang H, Li L, Wang S, Sun X, Luo C, Hou B. Construction of dentin-on-a-chip based on microfluidic technology and tissue engineering. J Dent 2024; 146:105028. [PMID: 38719135 DOI: 10.1016/j.jdent.2024.105028] [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: 02/05/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
AIM Three-dimensional (3D) cell culture systems perform better in resembling tissue or organism structures compared with traditional 2D models. Organs-on-chips (OoCs) are becoming more efficient 3D models. This study aimed to create a novel simplified dentin-on-a-chip using microfluidic chip technology and tissue engineering for screening dental materials. METHODOLOGY A microfluidic device with three channels was designed for creating 3D dental tissue constructs using stem cells from the apical papilla (SCAP) and gelatin methacrylate (GelMA). The study investigated the effect of varying cell densities and GelMA concentrations on the layer features formed within the microfluidic chip. Cell viability and distribution were evaluated through live/dead staining and nuclei/F-actin staining. The osteo/odontogenic potential was assessed through ALP staining and Alizarin red staining. The impact of GelMA concentrations (5 %, 10 %) on the osteo/odontogenic differentiation trajectory of SCAP was also studied. RESULTS The 3D tissue constructs maintained high viability and favorable spreading within the microfluidic chip for 3-7 days. A cell seeding density of 2 × 104 cells/μL was found to be the most optimal choice, ensuring favorable cell proliferation and even distribution. GelMA concentrations of 5 % and 10 % proved to be most effective for promoting cell growth and uniform distribution. Within the 5 % GelMA group, SCAP demonstrated higher osteo/odontogenic differentiation than that in the 10 % GelMA group. CONCLUSION In 3D culture, GelMA concentration was found to regulate the osteo/odontogenic differentiation of SCAP. The study recommends a seeding density of 2 × 104 cells/μL of SCAP within 5 % GelMA for constructing simplified dentin-on-a-chip. CLINICAL SIGNIFICANCE This study built up the 3D culture protocol, and induced odontogenic differentiation of SCAP, thus forming the simplified dentin-on-a-chip and paving the way to be used as a well-defined biological model for regenerative endodontics. It may serve as a potential testing platform for cell differentiation.
Collapse
Affiliation(s)
- Hexuan Zhang
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing 100162, PR China; Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing 100050, PR China
| | - Lingjun Li
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou 325001, PR China.
| | - Shujing Wang
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou 325001, PR China
| | - Xiaoqiang Sun
- Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing 100050, PR China
| | - Chunxiong Luo
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou 325001, PR China; The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, PR China.
| | - Benxiang Hou
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing 100162, PR China.
| |
Collapse
|
12
|
Sun M, Bai S, Wang H, Li Z, Wang Y, Guo X. Localized self-assembly of macroscopically structured supramolecular hydrogels through reaction-diffusion. SOFT MATTER 2024; 20:4776-4782. [PMID: 38842423 DOI: 10.1039/d4sm00467a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Localized molecular self-assembly has been developed as an effective approach for the fabrication of spatially resolved supramolecular hydrogels, showing great potential for many high-tech applications. However, the fabrication of macroscopically structured supramolecular hydrogels through molecular self-assembly remains a challenge. Herein, we report on localized self-assembly of low molecular weight hydrogelators through a simple reaction-diffusion approach, giving rise to various macroscopically patterned supramolecular hydrogels. This is achieved on the basis of an acid-catalyzed hydrazone supramolecular hydrogelator system. The acid was pre-loaded in a polydimethylsiloxane (PDMS) substrate, generating a proton gradient in the vicinity of the PDMS surface after immersing the PDMS in the aqueous solution of the hydrogelator precursors. The acid dramatically accelerates the in situ formation and self-assembly of the hydrazone hydrogelators, leading to localized formation of supramolecular hydrogels. The growth rate of the supramolecular hydrogels can be easily tuned through controlling the concentrations of the hydrogelator precursors and HCl. Importantly, differently shaped supramolecular hydrogel objects can be obtained by simply changing the shapes of PDMS. This work suggests that reaction-diffusion-mediated localized hydrogelation can serve as an approach towards macroscopically structuralized supramolecular hydrogels, which may find potential applications ranging from tissue engineering to biosensors.
Collapse
Affiliation(s)
- Mengran Sun
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Shengyu Bai
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Hucheng Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Zhongqi Li
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Yiming Wang
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai 200237, China.
| | - Xuhong Guo
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
13
|
Marinaro G, Bruno L, Pirillo N, Coluccio ML, Nanni M, Malara N, Battista E, Bruno G, De Angelis F, Cancedda L, Di Mascolo D, Gentile F. The role of elasticity on adhesion and clustering of neurons on soft surfaces. Commun Biol 2024; 7:617. [PMID: 38778159 PMCID: PMC11111731 DOI: 10.1038/s42003-024-06329-9] [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: 09/06/2023] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The question of whether material stiffness enhances cell adhesion and clustering is still open to debate. Results from the literature are seemingly contradictory, with some reports illustrating that adhesion increases with surface stiffness and others suggesting that the performance of a system of cells is curbed by high values of elasticity. To address the role of elasticity as a regulator in neuronal cell adhesion and clustering, we investigated the topological characteristics of networks of neurons on polydimethylsiloxane (PDMS) surfaces - with values of elasticity (E) varying in the 0.55-2.65 MPa range. Results illustrate that, as elasticity increases, the number of neurons adhering on the surface decreases. Notably, the small-world coefficient - a topological measure of networks - also decreases. Numerical simulations and functional multi-calcium imaging experiments further indicated that the activity of neuronal cells on soft surfaces improves for decreasing E. Experimental findings are supported by a mathematical model, that explains adhesion and clustering of cells on soft materials as a function of few parameters - including the Young's modulus and roughness of the material. Overall, results indicate that - in the considered elasticity interval - increasing the compliance of a material improves adhesion, improves clustering, and enhances communication of neurons.
Collapse
Affiliation(s)
- Giovanni Marinaro
- Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Quartier Hôpital, 4000, Liège, Belgium
| | - Luigi Bruno
- Department of Mechanical, Energy and Management Engineering, University of Calabria, 87036, Rende, Italy
| | - Noemi Pirillo
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University of "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Maria Laura Coluccio
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University of "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Marina Nanni
- Department of Neuroscience and Brain Technologies, Italian Institute of Technology, Via Morego 30, 16163, Genoa, Italy
| | - Natalia Malara
- Department of Health Science, University of "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Edmondo Battista
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Giulia Bruno
- Plasmon Nanotechnologies, Italian Institute of Technology, Via Morego 30, 16163, Genoa, Italy
| | - Francesco De Angelis
- Plasmon Nanotechnologies, Italian Institute of Technology, Via Morego 30, 16163, Genoa, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Italian Institute of Technology, Via Morego 30, 16163, Genoa, Italy
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, 16163, Genoa, Italy.
- Department of Electrical and Information Engineering, Polytechnic University of Bari, 70126, Bari, Italy.
| | - Francesco Gentile
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University of "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy.
| |
Collapse
|
14
|
Yu X, Park S, Lee S, Joo SW, Choo J. Microfluidics for disease diagnostics based on surface-enhanced raman scattering detection. NANO CONVERGENCE 2024; 11:17. [PMID: 38687445 PMCID: PMC11061072 DOI: 10.1186/s40580-024-00424-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/08/2024] [Indexed: 05/02/2024]
Abstract
This review reports diverse microfluidic systems utilizing surface-enhanced Raman scattering (SERS) detection for disease diagnosis. Integrating SERS detection technology, providing high-sensitivity detection, and microfluidic technology for manipulating small liquid samples in microdevices has expanded the analytical capabilities previously confined to larger settings. This study explores the principles and uses of various SERS-based microfluidic devices developed over the last two decades. Specifically, we investigate the operational principles of documented SERS-based microfluidic devices, including continuous-flow channels, microarray-embedded microfluidic channels, droplet microfluidic channels, digital droplet channels, and gradient microfluidic channels. We also examine their applications in biomedical diagnostics. In conclusion, we summarize the areas requiring further development to translate these SERS-based microfluidic technologies into practical applications in clinical diagnostics.
Collapse
Affiliation(s)
- Xiangdong Yu
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea
| | - Sohyun Park
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea
| | - Sungwoon Lee
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea
| | - Sang-Woo Joo
- Department of Chemistry, Soongsil University, Seoul, 06978, South Korea.
| | - Jaebum Choo
- Department of Chemistry, Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
15
|
Zhu C, Takemoto H, Higuchi Y, Yamashita F. Programmed immobilization of living cells using independent click pairs. Biochem Biophys Res Commun 2024; 699:149556. [PMID: 38277727 DOI: 10.1016/j.bbrc.2024.149556] [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: 10/10/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 01/28/2024]
Abstract
Therapeutic devices incorporating living cells or tissues have been intensively investigated for applications in tissue engineering and regenerative medicine. Because many biological processes are governed by spatially dependent signals, programmable immobilization of materials is crucial for manipulating multiple types of cells. In this study, click chemistry substrates were introduced onto the surfaces of cells and cover glass, and the cells were fixed on the cover glass via covalent bonds for selective cell deposition. Azide group (Az)-labeled living cells were prepared by metabolic labeling with azido sugars. Following the introduction of Az, TCO (trans-cyclooctene) was metabolically labeled into the living cells by reacting with TCO-DBCO (dibenzocyclooctyne). Az and TCO in the cells were detected using DBCO-FAM (fluorescein)and tetrazine-Cy3, respectively. The mixture of Az-labeled green fluorescent protein HeLa cells and TCO-labeled red fluorescent protein HeLa cells was reacted in a culture dish in which three different cover glasses, DBCO-, tetrazine-, or methyl-coated, were added. Az- or TCO-labeled cells could be immobilized in a functional group-dependent manner. Next, tetrazine-labeled cells were incubated on TCO- or Az-labeled cell layers instead of cover glass. Functional group-dependent immobilization was also achieved in the cell layer. Introducing substrates for the click reaction could achieve cell-selective immobilization on different patterned glass surfaces, as well as cell-cell immobilization.
Collapse
Affiliation(s)
- Chengyuan Zhu
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Hiroyasu Takemoto
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | - Yuriko Higuchi
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan; Department of Applied Pharmaceutics and Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
16
|
Kurian AG, Singh RK, Sagar V, Lee JH, Kim HW. Nanozyme-Engineered Hydrogels for Anti-Inflammation and Skin Regeneration. NANO-MICRO LETTERS 2024; 16:110. [PMID: 38321242 PMCID: PMC10847086 DOI: 10.1007/s40820-024-01323-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/24/2023] [Indexed: 02/08/2024]
Abstract
Inflammatory skin disorders can cause chronic scarring and functional impairments, posing a significant burden on patients and the healthcare system. Conventional therapies, such as corticosteroids and nonsteroidal anti-inflammatory drugs, are limited in efficacy and associated with adverse effects. Recently, nanozyme (NZ)-based hydrogels have shown great promise in addressing these challenges. NZ-based hydrogels possess unique therapeutic abilities by combining the therapeutic benefits of redox nanomaterials with enzymatic activity and the water-retaining capacity of hydrogels. The multifaceted therapeutic effects of these hydrogels include scavenging reactive oxygen species and other inflammatory mediators modulating immune responses toward a pro-regenerative environment and enhancing regenerative potential by triggering cell migration and differentiation. This review highlights the current state of the art in NZ-engineered hydrogels (NZ@hydrogels) for anti-inflammatory and skin regeneration applications. It also discusses the underlying chemo-mechano-biological mechanisms behind their effectiveness. Additionally, the challenges and future directions in this ground, particularly their clinical translation, are addressed. The insights provided in this review can aid in the design and engineering of novel NZ-based hydrogels, offering new possibilities for targeted and personalized skin-care therapies.
Collapse
Affiliation(s)
- Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Varsha Sagar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea.
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
17
|
Ran C, Pu K. Molecularly generated light and its biomedical applications. Angew Chem Int Ed Engl 2024; 63:e202314468. [PMID: 37955419 DOI: 10.1002/anie.202314468] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 11/14/2023]
Abstract
Molecularly generated light, referred to here as "molecular light", mainly includes bioluminescence, chemiluminescence, and Cerenkov luminescence. Molecular light possesses unique dual features of being both a molecule and a source of light. Its molecular nature enables it to be delivered as molecules to regions deep within the body, overcoming the limitations of natural sunlight and physically generated light sources like lasers and LEDs. Simultaneously, its light properties make it valuable for applications such as imaging, photodynamic therapy, photo-oxidative therapy, and photobiomodulation. In this review article, we provide an updated overview of the diverse applications of molecular light and discuss the strengths and weaknesses of molecular light across various domains. Lastly, we present forward-looking perspectives on the potential of molecular light in the realms of molecular imaging, photobiological mechanisms, therapeutic applications, and photobiomodulation. While some of these perspectives may be considered bold and contentious, our intent is to inspire further innovations in the field of molecular light applications.
Collapse
Affiliation(s)
- Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore, Singapore
| |
Collapse
|
18
|
Ouedraogo LJ, Trznadel MJ, Kling M, Nasirian V, Borst AG, Shirsavar MA, Makowski A, McNamara MC, Montazami R, Hashemi NN. Hydrodynamic Assembly of Astrocyte Cells in Conductive Hollow Microfibers. Adv Biol (Weinh) 2024; 8:e2300455. [PMID: 37953458 DOI: 10.1002/adbi.202300455] [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: 08/28/2023] [Revised: 10/19/2023] [Indexed: 11/14/2023]
Abstract
The manufacturing of 3D cell scaffoldings provides advantages for modeling diseases and injuries as it enables the creation of physiologically relevant platforms. A triple-flow microfluidic device is developed to rapidly fabricate alginate/graphene hollow microfibers based on the gelation of alginate induced with CaCl2 . This five-channel microdevice actualizes continuous mild fabrication of hollow fibers under an optimized flow rate ratio of 300:200:100 µL min-1 . The polymer solution is 2.5% alginate in 0.1% graphene and a 30% polyethylene glycol solution is used as the sheath and core solutions. The biocompatibility of these conductive microfibers by encapsulating mouse astrocyte cells (C8D1A) within the scaffolds is investigated. The cells can successfully survive both the manufacturing process and prolonged encapsulation for up to 8 days, where there is between 18-53% of live cells on both the alginate microfibers and alginate/graphene microfibers. These unique 3D hollow scaffolds can significantly enhance the available surface area for nutrient transport to the cells. In addition, these conductive hollow scaffolds illustrate unique advantages such as 0.728 cm3 gr-1 porosity and two times more electrical conductivity in comparison to alginate scaffolds. The results confirm the potential of these scaffolds as a microenvironment that supports cell growth.
Collapse
Affiliation(s)
- Lionel J Ouedraogo
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Mychal J Trznadel
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
| | - McKayla Kling
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
- Neuroscience Graduate Program, Iowa State University, Ames, IA, 50011, USA
| | - Vahid Nasirian
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Alexandra G Borst
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
- Neuroscience Graduate Program, Iowa State University, Ames, IA, 50011, USA
| | | | - Andrew Makowski
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Marilyn C McNamara
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Reza Montazami
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Nicole N Hashemi
- Department of Mechanical Engineering, Iowa State University, Ames, IA, 50011, USA
- Neuroscience Graduate Program, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
19
|
Park R, Kang MS, Heo G, Shin YC, Han DW, Hong SW. Regulated Behavior in Living Cells with Highly Aligned Configurations on Nanowrinkled Graphene Oxide Substrates: Deep Learning Based on Interplay of Cellular Contact Guidance. ACS NANO 2024; 18:1325-1344. [PMID: 38099607 DOI: 10.1021/acsnano.2c09815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Micro-/nanotopographical cues have emerged as a practical and promising strategy for controlling cell fate and reprogramming, which play a key role as biophysical regulators in diverse cellular processes and behaviors. Extracellular biophysical factors can trigger intracellular physiological signaling via mechanotransduction and promote cellular responses such as cell adhesion, migration, proliferation, gene/protein expression, and differentiation. Here, we engineered a highly ordered nanowrinkled graphene oxide (GO) surface via the mechanical deformation of an ultrathin GO film on an elastomeric substrate to observe specific cellular responses based on surface-mediated topographical cues. The ultrathin GO film on the uniaxially prestrained elastomeric substrate through self-assembly and subsequent compressive force produced GO nanowrinkles with periodic amplitude. To examine the acute cellular behaviors on the GO-based cell interface with nanostructured arrays of wrinkles, we cultured L929 fibroblasts and HT22 hippocampal neuronal cells. As a result, our developed cell-culture substrate obviously provided a directional guidance effect. In addition, based on the observed results, we adapted a deep learning (DL)-based data processing technique to precisely interpret the cell behaviors on the nanowrinkled GO surfaces. According to the learning/transfer learning protocol of the DL network, we detected cell boundaries, elongation, and orientation and quantitatively evaluated cell velocity, traveling distance, displacement, and orientation. The presented experimental results have intriguing implications such that the nanotopographical microenvironment could engineer the living cells' morphological polarization to assemble them into useful tissue chips consisting of multiple cell types.
Collapse
Affiliation(s)
- Rowoon Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Gyeonghwa Heo
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Yong Cheol Shin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio 44195, United States
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
20
|
Archana T, Nachammai N, Praveenkumar S. Optimizing Microfluidic Channel Design with High-Performance Materials for Safe Neonatal Drug Delivery. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2024; 18:294-303. [PMID: 39356100 DOI: 10.2174/0126673878292962240718055526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION Designing the microfluidic channel for neonatal drug delivery requires proper considerations to enhance the efficiency and safety of drug substances when used in neonates. Thus, this research aims to evaluate high-performance materials and optimize the channel design by modeling and simulation using COMSOL multiphysics in order to deliver an optimum flow rate between 0. 3 and 1 mL/hr. METHOD Some of the materials used in the study included PDMS, glass, COC, PMMA, PC, TPE, and hydrogels, and the evaluation criterion involved biocompatibility, mechanical properties, chemical resistance, and ease of fabrication. The simulation was carried out in the COMSOL multiphysics platform and demonstrated the fog fluid behavior in different channel geometries, including laminar flow and turbulence. The study then used systematic changes in design parameters with the aim of establishing the best implementation models that can improve the efficiency and reliability of the drug delivery system. The comparison was based mostly on each material and its appropriateness in microfluidic usage, primarily in neonatal drug delivery. The biocompatibility of the developed materials was verified using the literature analysis and adherence to the ISO 10993 standard, thus providing safety for the use of neonatal devices. Tensile strength was included to check the strength of each material to withstand its operation conditions. Chemical resistance was also tested in order to determine the compatibility of the materials with various drugs, and the possibility of fabrication was also taken into consideration to identify appropriate materials that could be used in the rapid manufacturing of the product. RESULTS The results we obtained show that PDMS, due to its flexibility and simplicity in simulation coupled with more efficient channel designs which have been extracted from COMSOL, present a feasible solution to neonatal drug delivery. CONCLUSION The present comparative study serves as a guide on the choice of materials and design of microfluidic devices to help achieve safer and enhanced drug delivery systems suitable for the delicate reception of fragile neonates.
Collapse
Affiliation(s)
- T Archana
- Department of Electronics and Instrumentation Engineering, Annamalai University, Annamalai Nagar, Chidambaram, India
| | - N Nachammai
- Department of Electronics and Instrumentation Engineering, Annamalai University, Annamalai Nagar, Chidambaram, India
| | - S Praveenkumar
- Department of Electronics and Communication Engineering, Saveetha Engineering College, Saveetha Nagar, Chennai, India
| |
Collapse
|
21
|
Wei SY, Chen PY, Hsieh CC, Chen YS, Chen TH, Yu YS, Tsai MC, Xie RH, Chen GY, Yin GC, Melero-Martin JM, Chen YC. Engineering large and geometrically controlled vascularized nerve tissue in collagen hydrogels to restore large-sized volumetric muscle loss. Biomaterials 2023; 303:122402. [PMID: 37988898 PMCID: PMC11606314 DOI: 10.1016/j.biomaterials.2023.122402] [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: 08/04/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Developing scalable vascularized and innervated tissue is a critical challenge for the successful clinical application of tissue-engineered constructs. Collagen hydrogels are extensively utilized in cell-mediated vascular network formation because of their naturally excellent biological properties. However, the substantial increase in hydrogel contraction induced by populated cells limits their long-term use. Previous studies attempted to mitigate this issue by concentrating collagen pre-polymer solutions or synthesizing covalently crosslinked collagen hydrogels. However, these methods only partially reduce hydrogel contraction while hindering blood vessel formation within the hydrogels. To address this challenge, we introduced additional support in the form of a supportive spacer to counteract the contraction forces of populated cells and prevent hydrogel contraction. This approach was found to promote cell spreading, resist hydrogel contraction, control hydrogel/tissue geometry, and even facilitate the engineering of functional blood vessels and host nerve growth in just one week. Subsequently, implanting these engineered tissues into muscle defect sites resulted in timely anastomosis with the host vasculature, leading to enhanced myogenesis, increased muscle innervation, and the restoration of injured muscle functionality. Overall, this innovative strategy expands the applicability of collagen hydrogels in fabricating large vascularized nerve tissue constructs for repairing volumetric muscle loss (∼63 %) and restoring muscle function.
Collapse
Affiliation(s)
- Shih-Yen Wei
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Po-Yu Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Chia-Chang Hsieh
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Yu-Shan Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Tzu-Hsuan Chen
- Department of Materials Science and Engineering, Carnegie Mellon University, PA, USA
| | - Yu-Shan Yu
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Chun Tsai
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Ren-Hao Xie
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Guan-Yu Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Gung-Chian Yin
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ying-Chieh Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan.
| |
Collapse
|
22
|
Zhang J, Liu C, Li X, Liu Z, Zhang Z. Application of photo-crosslinkable gelatin methacryloyl in wound healing. Front Bioeng Biotechnol 2023; 11:1303709. [PMID: 38076425 PMCID: PMC10702353 DOI: 10.3389/fbioe.2023.1303709] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/10/2023] [Indexed: 02/13/2025] Open
Abstract
Wound healing is a complex and coordinated biological process easily influenced by various internal and external factors. Hydrogels have immense practical importance in wound nursing because of their environmental moisturising, pain-relieving, and cooling effects. As photo-crosslinkable biomaterials, gelatine methacryloyl (GelMA) hydrogels exhibit substantial potential for tissue repair and reconstruction because of their tunable and beneficial properties. GelMA hydrogels have been extensively investigated as scaffolds for cell growth and drug release in various biomedical applications. They also hold great significance in wound healing because of their similarity to the components of the extracellular matrix of the skin and their favourable physicochemical properties. These hydrogels can promote wound healing and tissue remodelling by reducing inflammation, facilitating vascularisation, and supporting cell growth. In this study, we reviewed the applications of GelMA hydrogels in wound healing, including skin tissue engineering, wound dressing, and transdermal drug delivery. We aim to inspire further exploration of their potential for wound healing.
Collapse
Affiliation(s)
- Jinli Zhang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Changling Liu
- Department of Burns and Plastic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Xiaojian Li
- Department of Burns and Plastic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Zhihe Liu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Zhi Zhang
- Department of Burns and Plastic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| |
Collapse
|
23
|
Li D, Armand LC, Sun F, Hwang H, Wolfson D, Rampoldi A, Liu R, Forghani P, Hu X, Yu WM, Qu CK, Jones DP, Wu R, Cho HC, Maxwell JT, Xu C. AMPK activator-treated human cardiac spheres enhance maturation and enable pathological modeling. Stem Cell Res Ther 2023; 14:322. [PMID: 37941041 PMCID: PMC10633979 DOI: 10.1186/s13287-023-03554-7] [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: 02/21/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Cardiac pathological outcome of metabolic remodeling is difficult to model using cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) due to low metabolic maturation. METHODS hiPSC-CM spheres were treated with AMP-activated protein kinase (AMPK) activators and examined for hiPSC-CM maturation features, molecular changes and the response to pathological stimuli. RESULTS Treatment of hiPSC-CMs with AMPK activators increased ATP content, mitochondrial membrane potential and content, mitochondrial DNA, mitochondrial function and fatty acid uptake, indicating increased metabolic maturation. Conversely, the knockdown of AMPK inhibited mitochondrial maturation of hiPSC-CMs. In addition, AMPK activator-treated hiPSC-CMs had improved structural development and functional features-including enhanced Ca2+ transient kinetics and increased contraction. Transcriptomic, proteomic and metabolomic profiling identified differential levels of expression of genes, proteins and metabolites associated with a molecular signature of mature cardiomyocytes in AMPK activator-treated hiPSC-CMs. In response to pathological stimuli, AMPK activator-treated hiPSC-CMs had increased glycolysis, and other pathological outcomes compared to untreated cells. CONCLUSION AMPK activator-treated cardiac spheres could serve as a valuable model to gain novel insights into cardiac diseases.
Collapse
Affiliation(s)
- Dong Li
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Lawrence C Armand
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Fangxu Sun
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hyun Hwang
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - David Wolfson
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Antonio Rampoldi
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Rui Liu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Xin Hu
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Wen-Mei Yu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Dean P Jones
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hee Cheol Cho
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Joshua T Maxwell
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
24
|
Samal P, Kumar Samal JR, Rho HS, van Beurden D, van Blitterswijk C, Truckenmüller R, Giselbrecht S. Direct deep UV lithography to micropattern PMMA for stem cell culture. Mater Today Bio 2023; 22:100779. [PMID: 37701129 PMCID: PMC10494264 DOI: 10.1016/j.mtbio.2023.100779] [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: 03/17/2023] [Revised: 06/22/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023] Open
Abstract
Microengineering is increasingly being used for controlling the microenvironment of stem cells. Here, a novel method for fabricating structures with subcellular dimensions in commonly available thermoplastic poly(methyl methacrylate) (PMMA) is shown. Microstructures are produced in PMMA substrates using Deep Ultraviolet lithography, and the effect of different developers is described. Microgrooves fabricated in PMMA are used for the neuronal differentiation of mouse embryonic stem cells (mESCs) directly on the polymer. The fabrication of 3D, curvilinear patterned surfaces is also highlighted. A 3D multilayered microfluidic chip is fabricated using this method, which includes a porous polycarbonate (PC) membrane as cell culture substrate. Besides directly manufacturing PMMA-based microfluidic devices, an application of the novel approach is shown where a reusable PMMA master is created for replicating microstructures with polydimethylsiloxane (PDMS). As an application example, microchannels fabricated in PDMS are used to selectively expose mESCs to soluble factors in a localized manner. The described microfabrication process offers a remarkably simple method to fabricate for example multifunctional topographical or microfluidic culture substrates outside cleanrooms, thereby using inexpensive and widely accessible equipment. The versatility of the underlying process could find various applications also in optical systems and surface modification of biomedical implants.
Collapse
Affiliation(s)
- Pinak Samal
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Jay Rabindra Kumar Samal
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Hoon Suk Rho
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
- Mepsgen Co., Ltd., 7F, Hanyang Tower, 12, Beobwon-ro-11-gil, Songpa-gu, Seoul, Republic of Korea
| | - Denis van Beurden
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Clemens van Blitterswijk
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
25
|
Luo K, Wang L, Wang MX, Du R, Tang L, Yang KK, Wang YZ. 4D Printing of Biocompatible Scaffolds via In Situ Photo-crosslinking from Shape Memory Copolyesters. ACS APPLIED MATERIALS & INTERFACES 2023; 15:44373-44383. [PMID: 37669475 DOI: 10.1021/acsami.3c10747] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
The complexity of surgical treatments for large-area soft tissue injuries makes placing large implants into injury sites challenging. Aliphatic polyesters are often used for scaffold preparation in tissue engineering owing to their excellent biodegradability and biocompatibility. Scaffolds with shape-memory effect (SME) can also avoid large-volume trauma during the implantation. However, the complexity and diversity of diseases require more adaptable and precise processing methods. Four-dimensional (4D) printing, a booming smart material additive manufacturing technology, provides a new opportunity for developing shape memory scaffolds. With the aim of personalized or patient-adaptable soft tissues such as blood vessels, we developed a feasible strategy for fabricating scaffolds with fine architectures using 4D printing crosslinkable shape memory linear copolyesters using fused deposition modeling (FDM). To overcome the weak bonding strength of each printed layer during FDM, a catalyst-free photo-crosslinkable functional group derived from biocompatible cinnamic acid was embedded into the linear copolyesters as in situ crosslinking points during FDM printing. Under ultraviolet-assisted irradiation, the resulting 4D scaffold models demonstrated excellent SME, desirable mechanical performance, and good stability in a water environment owing to the chemical bonding between each layer. Moreover, the excellent biocompatibility of the scaffold was evaluated in vitro and in vivo. The developed composite scaffolds could be used for minimally invasive soft tissue repair.
Collapse
Affiliation(s)
- Kun Luo
- Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Li Wang
- Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), College of Chemistry, Sichuan University, Chengdu 610064, China
- Department of Biomedical Engineering, School of Big Health and Intelligent Engineering, Chengdu 610500, China
| | - Man-Xi Wang
- Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Rui Du
- Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Li Tang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke-Ke Yang
- Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yu-Zhong Wang
- Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), State Key Laboratory of Polymer Materials Engineering, National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), College of Chemistry, Sichuan University, Chengdu 610064, China
| |
Collapse
|
26
|
Liu D, Meng Q, Hu J. Bacterial Nanocellulose Hydrogel: A Promising Alternative Material for the Fabrication of Engineered Vascular Grafts. Polymers (Basel) 2023; 15:3812. [PMID: 37765666 PMCID: PMC10534661 DOI: 10.3390/polym15183812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
Blood vessels are crucial in the human body, providing essential nutrients to all tissues while facilitating waste removal. As the incidence of cardiovascular disease rises, the demand for efficient treatments increases concurrently. Currently, the predominant interventions for cardiovascular disease are autografts and allografts. Although effective, they present limitations including high costs and inconsistent success rates. Recently, synthetic vascular grafts, made from artificial materials, have emerged as promising alternatives to traditional methods. Among these materials, bacterial cellulose hydrogel exhibits significant potential for tissue engineering applications, particularly in developing nanoscale platforms that regulate cell behavior and promote tissue regeneration, attributed to its notable physicochemical and biocompatible properties. This study reviews recent progress in fabricating engineered vascular grafts using bacterial nanocellulose, demonstrating the efficacy of bacterial cellulose hydrogel as a biomaterial for synthetic vascular grafts, specifically for stimulating angiogenesis and neovascularization.
Collapse
Affiliation(s)
| | | | - Jinguang Hu
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive, Calgary, AB T2N 1N4, Canada; (D.L.); (Q.M.)
| |
Collapse
|
27
|
Nie L, Liu W, Chen J, Zhou S, Liu C, Li W, Ran Z, Liu Y, Hu J, Zhang Y, Zheng L, Ji P, Zhang H. A Novel Bioimplant Comprising Ad-BMP9-Transfected BMSCs and GelMA Microspheres Produced from Microfluidic Devices for Bone Tissue Engineering. J Tissue Eng Regen Med 2023; 2023:2981936. [PMID: 40226408 PMCID: PMC11918572 DOI: 10.1155/2023/2981936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/30/2023] [Accepted: 06/05/2023] [Indexed: 04/15/2025]
Abstract
Oral and maxillofacial bone defect repair in patients remains challenging in clinical treatment due to the different morphologies of bone defects. An injectable hydrogel of microspheres with sustained bone morphogenetic protein 9 (BMP9) expression for oral and maxillofacial bone defect repair has been developed. This study is bioinspired by the substantial osteogenesis property of recombinant adenoviruses expressing bone morphogenetic protein 9 (Ad-BMP9) and minimally invasive treatment by injection. A novel scaffold encompassing bone mesenchymal stem cells (BMSCs) transfected with Ad-BMP9 was produced and cocultured on a superficial surface of monodisperse photocrosslinked methacrylate gelatin hydrogel microspheres (GelMA/MS, produced with microfluidic technology). The biological tests including live/dead cell staining, phalloidin staining, cell counting kit-8 (CCK-8) assay, alkaline phosphatase (ALP) activity and staining, alizarin red S staining, and quantitative real-time polymerase chain reaction (RT-qPCR), revealed that the hydrogel microspheres exhibited good biocompatibility and remarkably promoted the osteogenic differentiation of BMSCs in vitro. In addition, a small needle was injected the innovative scaffold beneath the nude mice's skin. The micro-CT and histological staining assay results demonstrated that the new implant, with high blood vessel formation markers (CD31-positive cells) expression over four and eight weeks, achieved significant vascularized bone-like tissue formation. Consequently, the injectable hydrogel microspheres, cocultured with BMSC transfected with Ad-BMP9, enhanced vascularized bone regeneration, therefore representing a facile and promising technique for the minimally invasive treatment of oral and maxillofacial bone defects.
Collapse
Affiliation(s)
- Li Nie
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Wei Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Jiajun Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Siqi Zhou
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Chang Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Wenhui Li
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Zhiyue Ran
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Yaxian Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Jing Hu
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Yuxin Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Liwen Zheng
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Hongmei Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key, Chongqing 401147, China
- Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| |
Collapse
|
28
|
Chu PY, Hsieh HY, Chung PS, Wang PW, Wu MC, Chen YQ, Kuo JC, Fan YJ. Development of vessel mimicking microfluidic device for studying mechano-response of endothelial cells. iScience 2023; 26:106927. [PMID: 37305698 PMCID: PMC10251125 DOI: 10.1016/j.isci.2023.106927] [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: 12/21/2021] [Revised: 10/24/2022] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
The objective of this study is to develop a device to mimic a microfluidic system of human arterial blood vessels. The device combines fluid shear stress (FSS) and cyclic stretch (CS), which are resulting from blood flow and blood pressure, respectively. The device can reveal real-time observation of dynamic morphological change of cells in different flow fields (continuous flow, reciprocating flow and pulsatile flow) and stretch. We observe the effects of FSS and CS on endothelial cells (ECs), including ECs align their cytoskeleton proteins with the fluid flow direction and paxillin redistribution to the cell periphery or the end of stress fibers. Thus, understanding the morphological and functional changes of endothelial cells on physical stimuli can help us to prevent and improve the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Pei-Yu Chu
- College of Biomedical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Han-Yun Hsieh
- College of Biomedical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Pei-Shan Chung
- Department of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Pai-Wen Wang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 11221, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 11221, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 11221, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 11221, Taiwan
| | - Yu-Jui Fan
- College of Biomedical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
- International Ph.D. Program for Biomedical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| |
Collapse
|
29
|
Lee JC, Brien HJ, Walton BL, Eidman ZM, Toda S, Lim WA, Brunger JM. Instructional materials that control cellular activity through synthetic Notch receptors. Biomaterials 2023; 297:122099. [PMID: 37023529 PMCID: PMC10320837 DOI: 10.1016/j.biomaterials.2023.122099] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
The field of regenerative engineering relies primarily on the dual technical platforms of cell selection/conditioning and biomaterial fabrication to support directed cell differentiation. As the field has matured, an appreciation for the influence of biomaterials on cell behaviors has resulted in engineered matrices that meet biomechanical and biochemical demands of target pathologies. Yet, despite advances in methods to produce designer matrices, regenerative engineers remain unable to reliably orchestrate behaviors of therapeutic cells in situ. Here, we present a platform named MATRIX whereby cellular responses to biomaterials can be custom defined by combining engineered materials with cells expressing cognate synthetic biology control modules. Such privileged channels of material-to-cell communication can activate synthetic Notch receptors and govern activities as diverse as transcriptome engineering, inflammation attenuation, and pluripotent stem cell differentiation, all in response to materials decorated with otherwise bioinert ligands. Further, we show that engineered cellular behaviors are confined to programmed biomaterial surfaces, highlighting the potential to use this platform to spatially organize cellular responses to bulk, soluble factors. This integrated approach of co-engineering cells and biomaterials for orthogonal interactions opens new avenues for reproducible control of cell-based therapies and tissue replacements.
Collapse
Affiliation(s)
- Joanne C Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Hannah J Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Zachary M Eidman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Satoshi Toda
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Wendell A Lim
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA.
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA.
| |
Collapse
|
30
|
Wang R, Wang M, Jin R, Wang Y, Yi M, Li Q, Li J, Zhang K, Sun C, Nie Y, Huang C, Mikos AG, Zhang X. High Strength Titanium with Fibrous Grain for Advanced Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207698. [PMID: 37029460 PMCID: PMC10238201 DOI: 10.1002/advs.202207698] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/21/2023] [Indexed: 06/04/2023]
Abstract
Pure titanium is widely used in clinical implants, but its bioinert properties (poor strength and mediocre effect on bone healing) limit its use under load-bearing conditions. Modeling on the structure of collagen fibrils and specific nanocrystal plane arrangement of hydroxyapatite in the natural bone, a new type of titanium (Ti) with a highly aligned fibrous-grained (FG) microstructure is constructed. The improved attributes of FG Ti include high strength (≈950 MPa), outstanding affinity to new bone growth, and tight bone-implant contact. The bone-mimicking fibrous grains induce an aligned surface topological structure conducive to forming close contact with osteoblasts and promotes the expression of osteogenic genes. Concurrently, the predominant Ti(0002) crystal plane of FG Ti induces the formation of hydrophilic anatase titanium oxide layers, which accelerate biomineralization. In conclusion, this bioinspired FG Ti not only proves to show mechanical and bone-regenerative improvements but it also provides a new strategy for the future design of metallic biomaterials.
Collapse
Affiliation(s)
- Ruohan Wang
- National Engineering Research Centre for Biomaterials/College of Biomedical EngineeringSichuan UniversityChengdu610065China
| | - Mingsai Wang
- School of Aeronautics and AstronauticsSichuan UniversityChengdu610065China
| | - Rongrong Jin
- National Engineering Research Centre for Biomaterials/College of Biomedical EngineeringSichuan UniversityChengdu610065China
| | - Yanfei Wang
- School of Aeronautics and AstronauticsSichuan UniversityChengdu610065China
| | - Min Yi
- Department of OrthopedicsOrthopedic Research InstituteWest China HospitalSichuan UniversityChengdu610041China
| | - Qinye Li
- Department of Chemistry and BiotechnologyCentre for Translational AtomaterialsSwinburne University of TechnologyHawthornVIC3122Australia
| | - Juan Li
- State Key Laboratory of Oral DiseasesWest China School of StomatologyWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Kai Zhang
- National Engineering Research Centre for Biomaterials/College of Biomedical EngineeringSichuan UniversityChengdu610065China
| | - Chenghua Sun
- Department of Chemistry and BiotechnologyCentre for Translational AtomaterialsSwinburne University of TechnologyHawthornVIC3122Australia
| | - Yu Nie
- National Engineering Research Centre for Biomaterials/College of Biomedical EngineeringSichuan UniversityChengdu610065China
| | - Chongxiang Huang
- National Engineering Research Centre for Biomaterials/College of Biomedical EngineeringSichuan UniversityChengdu610065China
- School of Aeronautics and AstronauticsSichuan UniversityChengdu610065China
| | - Antonios G. Mikos
- Departments of BioengineeringChemical and Biomolecular EngineeringRice UniversityHoustonTX77251USA
| | - Xingdong Zhang
- National Engineering Research Centre for Biomaterials/College of Biomedical EngineeringSichuan UniversityChengdu610065China
| |
Collapse
|
31
|
Liu S, Yu JM, Gan YC, Qiu XZ, Gao ZC, Wang H, Chen SX, Xiong Y, Liu GH, Lin SE, McCarthy A, John JV, Wei DX, Hou HH. Biomimetic natural biomaterials for tissue engineering and regenerative medicine: new biosynthesis methods, recent advances, and emerging applications. Mil Med Res 2023; 10:16. [PMID: 36978167 PMCID: PMC10047482 DOI: 10.1186/s40779-023-00448-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Biomimetic materials have emerged as attractive and competitive alternatives for tissue engineering (TE) and regenerative medicine. In contrast to conventional biomaterials or synthetic materials, biomimetic scaffolds based on natural biomaterial can offer cells a broad spectrum of biochemical and biophysical cues that mimic the in vivo extracellular matrix (ECM). Additionally, such materials have mechanical adaptability, microstructure interconnectivity, and inherent bioactivity, making them ideal for the design of living implants for specific applications in TE and regenerative medicine. This paper provides an overview for recent progress of biomimetic natural biomaterials (BNBMs), including advances in their preparation, functionality, potential applications and future challenges. We highlight recent advances in the fabrication of BNBMs and outline general strategies for functionalizing and tailoring the BNBMs with various biological and physicochemical characteristics of native ECM. Moreover, we offer an overview of recent key advances in the functionalization and applications of versatile BNBMs for TE applications. Finally, we conclude by offering our perspective on open challenges and future developments in this rapidly-evolving field.
Collapse
Affiliation(s)
- Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| | - Jiang-Ming Yu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336 China
| | - Yan-Chang Gan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| | - Xiao-Zhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| | - Zhe-Chen Gao
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336 China
| | - Huan Wang
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033 Guangdong China
| | - Shi-Xuan Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325011 Zhejiang China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Si-En Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077 China
| | - Alec McCarthy
- Department of Functional Materials, Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Johnson V. John
- Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68130 USA
| | - Dai-Xu Wei
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336 China
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong, 643002 Sichuan China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710127 China
| | - Hong-Hao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| |
Collapse
|
32
|
Che B, Sun D, Zhang C, Hou J, Zhao W, Jing G, Mu Y, Cao Y, Dai L, Zhang C. Gradient Nanoconfinement Facilitates Binding of Transcriptional Factor NF-κB to Histone- and Protamine-DNA Complexes. NANO LETTERS 2023; 23:2388-2396. [PMID: 36857512 DOI: 10.1021/acs.nanolett.3c00325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Mechanically induced chromosome reorganization plays important roles in transcriptional regulation. However, the interplay between chromosome reorganization and transcription activities is complicated, such that it is difficult to decipher the regulatory effects of intranuclear geometrical cues. Here, we simplify the system by introducing DNA, packaging proteins (i.e., histone and protamine), and transcription factor NF-κB into a well-defined fluidic chip with changing spatical confinement ranging from 100 to 500 nm. It is uncovered that strong nanoconfinement suppresses higher-order folding of histone- and protamine-DNA complexes, the fracture of which exposes buried DNA segments and causes increased quantities of NF-κB binding to the DNA chain. Overall, these results reveal a pathway of how intranuclear geometrical cues alter the open/closed state of a DNA-protein complex and therefore affect transcription activities: i.e., NF-κB binding.
Collapse
Affiliation(s)
- Bingchen Che
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an 710069, People's Republic of China
- School of Physics, Northwest University, Xi'an 710069, People's Republic of China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an 710069, People's Republic of China
| | - Chen Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an 710069, People's Republic of China
| | - Jiaqing Hou
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an 710069, People's Republic of China
| | - Wei Zhao
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an 710069, People's Republic of China
| | - Guangyin Jing
- School of Physics, Northwest University, Xi'an 710069, People's Republic of China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 639798, Singapore
| | - Yaoyu Cao
- Institute of Photonics Technology, Jinan University, 510632, Guangzhou, People's Republic of China
| | - Liang Dai
- Department of Physics, City University of Hong Kong, Hong Kong 999077, People's Republic of China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, People's Republic of China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon-Technology, Northwest University, Xi'an 710069, People's Republic of China
| |
Collapse
|
33
|
Treherne JM, Miller AF. Novel hydrogels: are they poised to transform 3D cell-based assay systems in early drug discovery? Expert Opin Drug Discov 2023; 18:335-346. [PMID: 36722285 DOI: 10.1080/17460441.2023.2175813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Success in drug discovery remains unpredictable. However, more predictive and relevant disease models are becoming pivotal to demonstrating the clinical benefits of new drugs earlier in the lengthy drug discovery process. Novel hydrogel scaffolds are being developed to transform the relevance of such 3D cell-based in vitro assay systems. AREAS COVERED Most traditional hydrogels are still of unknown composition and suffer significant batch-to-batch variations, which lead to technical constraints. This article looks at how a new generation of novel synthetic hydrogels that are based on self-assembling peptides are poised to transform 3D cell-based assay systems by improving their relevance, reproducibility and scalability. EXPERT OPINION The emerging advantages of using these novel hydrogels for human 3D screening assays should enable the discovery of more cost-effective drugs, leading to improved patient benefits. Such a disruptive change could also reduce the considerable time lag from obtaining in vitro assay data to initiating clinical trials. There is now a sufficient body of data available in the literature to enable this ambition to become a reality by significantly improving the predictive validity of 3D cell-based assays in early drug discovery. Novel hydrogels are key to unlocking the full potential of these assay systems.
Collapse
Affiliation(s)
- J Mark Treherne
- Talisman Therapeutics Ltd, Jonas Webb Building and Cell Guidance Sysyems Ltd, Babraham Research Campus, Cambridge, UK
| | - Aline F Miller
- Manchester Institute of Biotechnology, School of Engineering, The University of Manchester, Oxford Road, Manchester, UK
| |
Collapse
|
34
|
Kankala RK, Zhang YS, Kang L, Ambrosio L. Editorial: Polymeric microarchitectures for tissue regeneration and drug screening. Front Bioeng Biotechnol 2023; 11:1144991. [PMID: 36815895 PMCID: PMC9932963 DOI: 10.3389/fbioe.2023.1144991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Affiliation(s)
- Ranjith Kumar Kankala
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China,*Correspondence: Ranjith Kumar Kankala,
| | | | - Lifeng Kang
- Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| | | |
Collapse
|
35
|
Duan C, Yu M, Hu C, Xia H, Kankala RK. Polymeric microcarriers for minimally-invasive cell delivery. Front Bioeng Biotechnol 2023; 11:1076179. [PMID: 36777246 PMCID: PMC9908582 DOI: 10.3389/fbioe.2023.1076179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Tissue engineering (TE) aims at restoring tissue defects by applying the three-dimensional (3D) biomimetic pre-formed scaffolds to restore, maintain, and enhance tissue growth. Broadly speaking, this approach has created a potential impact in anticipating organ-building, which could reduce the need for organ replacement therapy. However, the implantation of such cell-laden biomimetic constructs based on substantial open surgeries often results in severe inflammatory reactions at the incision site, leading to the generation of a harsh adverse environment where cell survival is low. To overcome such limitations, micro-sized injectable modularized units based on various biofabrication approaches as ideal delivery vehicles for cells and various growth factors have garnered compelling interest owing to their minimally-invasive nature, ease of packing cells, and improved cell retention efficacy. Several advancements have been made in fabricating various 3D biomimetic microscale carriers for cell delivery applications. In this review, we explicitly discuss the progress of the microscale cell carriers that potentially pushed the borders of TE, highlighting their design, ability to deliver cells and substantial tissue growth in situ and in vivo from different viewpoints of materials chemistry and biology. Finally, we summarize the perspectives highlighting current challenges and expanding opportunities of these innovative carriers.
Collapse
Affiliation(s)
- Chunyan Duan
- School of New Energy and Environmental Protection Engineering, Foshan Polytechnic, Foshan, China,*Correspondence: Ranjith Kumar Kankala, ; Chunyan Duan,
| | - Mingjia Yu
- School of New Energy and Environmental Protection Engineering, Foshan Polytechnic, Foshan, China
| | - Changji Hu
- School of New Energy and Environmental Protection Engineering, Foshan Polytechnic, Foshan, China
| | - Hongying Xia
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Ranjith Kumar Kankala
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, China,*Correspondence: Ranjith Kumar Kankala, ; Chunyan Duan,
| |
Collapse
|
36
|
Pan H, Mei D, Xu C, Han S, Wang Y. Bisymmetric coherent acoustic tweezers based on modulation of surface acoustic waves for dynamic and reconfigurable cluster manipulation of particles and cells. LAB ON A CHIP 2023; 23:215-228. [PMID: 36420975 DOI: 10.1039/d2lc00812b] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Acoustic tweezers based on surface acoustic waves (SAWs) have raised great interest in the fields of tissue engineering, targeted therapy, and drug delivery. Generally, the complex structure and array layout design of interdigital electrodes would restrict the applications of acoustic tweezers. Here, we present a novel approach by using bisymmetric coherent acoustic tweezers to modulate the shape of acoustic pressure fields with high flexibility and accuracy. Experimental tests were conducted to perform the precise, contactless, and biocompatible cluster manipulation of polystyrene microparticles and yeast cells. Stripe, dot, quadratic lattice, hexagonal lattice, interleaved stripe, oblique stripe, and many other complex arrays were achieved by real-time modulation of amplitudes and phase relations of coherent SAWs to demonstrate the capability of the device for the cluster manipulation of particles and cells. Furthermore, rapid switching among various arrays, shape regulation, geometric parameter modulation of array units, and directional translation of microparticles and cells were implemented. This study demonstrated a favorable technique for flexible and versatile manipulation and patterning of cells and biomolecules, and it has the advantages of high manipulation accuracy and adjustability, thus it is expected to be utilized in the fields of targeted cellular assembly, biological 3D printing, and targeted release of drugs.
Collapse
Affiliation(s)
- Hemin Pan
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Deqing Mei
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Chengyao Xu
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Shuo Han
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yancheng Wang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.
| |
Collapse
|
37
|
de Silva L, Bernal PN, Rosenberg A, Malda J, Levato R, Gawlitta D. Biofabricating the vascular tree in engineered bone tissue. Acta Biomater 2023; 156:250-268. [PMID: 36041651 DOI: 10.1016/j.actbio.2022.08.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
The development of tissue engineering strategies for treatment of large bone defects has become increasingly relevant, given the growing demand for bone substitutes. Native bone is composed of a dense vascular network necessary for the regulation of bone development, regeneration and homeostasis. A major obstacle in fabricating living, clinically relevant-sized bone mimics (1-10 cm3) is the limited supply of nutrients, including oxygen to the core of the construct. Therefore, strategies to support vascularization are pivotal for the development of tissue engineered bone constructs. Creating a functional bone construct integrated with a vascular network, capable of delivering the necessary nutrients for optimal tissue development is imperative for translation into the clinics. The vascular system is composed of a complex network that runs throughout the body in a tree-like hierarchical branching fashion. A significant challenge for tissue engineering approaches lies in mimicking the intricate, multi-scale structures consisting of larger vessels (macro-vessels) which interconnect with multiple sprouting vessels (microvessels) in a closed network. The advent of biofabrication has enabled complex, out of plane channels to be generated and has laid the groundwork for the creation of multi-scale vasculature in recent years. This review highlights the key state-of-the-art achievements for the development of vascular networks of varying scales in the field of biofabrication with a particular focus for its application in developing a functional tissue engineered bone construct. STATEMENT OF SIGNIFICANCE: There is a growing need for bone substitutes to overcome the limited supply of patient-derived bone. Bone tissue engineering aims to overcome this by combining stem cells with scaffolds to restore missing bone. The current bottleneck in upscaling is the lack of an integrated vascular network, required for the delivery of nutrients to cells. 3D bioprinting techniques has enabled the creation of complex hollow structures of varying dimensions that resemble native blood vessels. The convergence of multiple materials, cell types and fabrication approaches, opens the possibility of developing clinically-relevant sized vascularized bone constructs. This review provides an up-to-date insight of the technologies currently available for the generation of complex vascular networks, with a focus on their application in bone tissue engineering.
Collapse
Affiliation(s)
- Leanne de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands.
| | - Paulina N Bernal
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Ajw Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands
| |
Collapse
|
38
|
Tayanloo-Beik A, Nikkhah A, Roudsari PP, Aghayan H, Rezaei-Tavirani M, Nasli-Esfahani E, Mafi AR, Nikandish M, Shouroki FF, Arjmand B, Larijani B. Application of Biocompatible Scaffolds in Stem-Cell-Based Dental Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:83-110. [PMID: 35999347 DOI: 10.1007/5584_2022_734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Tissue engineering as an important field in regenerative medicine is a promising therapeutic approach to replace or regenerate injured tissues. It consists of three vital steps including the selection of suitable cells, formation of 3d scaffolds, and adding growth factors. Mesenchymal stem cells (MSCs) and embryonic stem cells (ESCs) are mentioned as two main sources for this approach that have been used for the treatment of various types of disorders. However, the main focus of literature in the field of dental tissue engineering is on utilizing MSCs. On the other hand, biocompatible scaffolds play a notable role in this regenerative process which is mentioned to be harmless with acceptable osteoinductivity. Their ability in inhibiting inflammatory responses also makes them powerful tools. Indeed, stem cell functions should be supported by biomaterials acting as scaffolds incorporated with biological signals. Naturally derived polymeric scaffolds and synthetically engineered polymeric/ceramic scaffolds are two main types of scaffolds regarding their materials that are defined further in this review. Various strategies of tissue bioengineering can affect the regeneration of dentin-pulp complex, periodontium regeneration, and whole teeth bioengineering. In this regard, in vivo/ex vivo experimental models have been developed recently in order to perform preclinical studies of dental tissue engineering which make it more transferable to be used for clinic uses. This review summarizes dental tissue engineering through its different components. Also, strategies of tissue bioengineering and experimental models are introduced in order to provide a perspective of the potential roles of dental tissue engineering to be used for clinical aims.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirabbas Nikkhah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyvand Parhizkar Roudsari
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ensieh Nasli-Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Rezazadeh Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shaheed Beheshti Medical University, Tehran, Iran
| | - Mohsen Nikandish
- AJA Cancer Epidemiology Research and Treatment Center (AJA- CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fazeli Shouroki
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Tavasoli H, Tavasoli N, Yunessnia lehi A. Nanonodular porous membranes: Novel scaffolds for development and proliferation of dental pulp‐derived mesenchymal stem cells. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hanieh Tavasoli
- Faculty of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Nafiseh Tavasoli
- Faculty of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | | |
Collapse
|
40
|
Naveen NR, Girirajasekhar D, Goudanavar PS, Kumar CB, Narasimha GL. Prospection of Microfluidics for Local Drug Delivery. Curr Drug Targets 2022; 23:1239-1251. [PMID: 35379132 DOI: 10.2174/1389450123666220404154710] [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: 08/16/2021] [Revised: 01/03/2022] [Accepted: 02/10/2022] [Indexed: 01/25/2023]
Abstract
Significant endeavors can be made to develop effective drug delivery systems. Nowadays, many of these novel systems have gained attention as they focus primarily on increasing the bioavailability and bioaccessibility of several drugs to finally minimize the side effects, thus improving the treatment's efficacy. Microfluidics systems are unquestionably a superior technology, which is currently revolutionizing the current chemical and biological studies, providing diminutive chip-scale devices that offer precise dosage, target-precise delivery, and controlled release. Microfluidic systems have emerged as a promising delivery vehicle owing to their potential for defined handling and transporting of small liquid quantities. The latest microfabrication developments have been made for application to several biological systems. Here, we review the fundamentals of microfluidics and their application for local drug delivery.
Collapse
Affiliation(s)
- Nimbagal R Naveen
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B.G. Nagar, Karnataka 571448, India
| | | | - Prakash S Goudanavar
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B.G. Nagar, Karnataka 571448, India
| | - Chagaleti B Kumar
- Department of Pharmaceutical Chemistry, Akshaya Institute of Pharmacy, Lingapura, Tumkur, Karnataka 572106, India
| | - Gunturu L Narasimha
- Department of Pharmacy Practice, Annamacharya College of Pharmacy, New Boyanapalli, Rajampet, India
| |
Collapse
|
41
|
Patrick MD, Keys JF, Suresh Kumar H, Annamalai RT. Injectable nanoporous microgels generate vascularized constructs and support bone regeneration in critical-sized defects. Sci Rep 2022; 12:15811. [PMID: 36138042 PMCID: PMC9499928 DOI: 10.1038/s41598-022-19968-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022] Open
Abstract
Large and aberrant bone fractures require ossification and concomitant vascularization for proper healing. Evidence indicates that osteogenesis and vessel growth are coupled in bone fractures. Although the synergistic role of endothelial cells has been recognized, vascularizing large bone grafts remains a challenge and has apprehended the clinical translation of engineered bone constructs. Here, we describe a facile method to fabricate vascularized constructs using chitosan and gelatin-based microgels that promote osteogenesis of human mesenchymal stromal cells (MSC) while supporting endothelial sprouting and network formation. The microgels are enzymatically degradable and had a high hydration rate with a volume swelling ratio of ~ 493% and a polymer density of ~ 431 mg/cm3, which is comparable to that of native skeletal tissues. AFM indentation of the surface showed an average Young's modulus of 189 kPa, falling in a range that is conducive to both osteogenesis and vasculogenesis. The osteogenic microgel containing chitosan, gelatin, and hydroxyapatite, mimicking the bone matrix, supported robust attachment, proliferation, and differentiation of MSC. On the other hand, the vasculogenic microgels containing only gelatin, enriched endothelial phenotype and enabled vascular networks formation when embedded in 3D matrices. Combining the two types of microgels created a hybrid construct that sustained the functions of both osteogenic and vasculogenic microgels and enhanced one another. Using a murine model, we also show that the osteogenic microgels regenerate bone in a critical-sized defect with > 95% defect closure by week 12. These multifunctional microgels can be administered minimally invasively and can conformally fill large bone defects. This work lays the foundation to establish principles of designing multiphasic scaffolds with tissue-specific biophysical and biochemical properties for regenerating vascularized and interfacial tissues.
Collapse
Affiliation(s)
- Matthew D Patrick
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA
| | - Jeremy F Keys
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA
| | - Harshini Suresh Kumar
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA
| | - Ramkumar T Annamalai
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA.
| |
Collapse
|
42
|
Kim JY, Choi W, Mangal U, Seo JY, Kang TY, Lee J, Kim T, Cha JY, Lee KJ, Kim KM, Kim JM, Kim D, Kwon JS, Hong J, Choi SH. Multivalent network modifier upregulates bioactivity of multispecies biofilm-resistant polyalkenoate cement. Bioact Mater 2022; 14:219-233. [PMID: 35310353 PMCID: PMC8897648 DOI: 10.1016/j.bioactmat.2021.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/25/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Affiliation(s)
- Ji-Yeong Kim
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Woojin Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Utkarsh Mangal
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ji-Young Seo
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Tae-Yun Kang
- Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Joohee Lee
- Johns Hopkins University, 3400 N. Charles St., Mason Hall, Baltimore, MD 21218, USA
| | - Taeho Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jung-Yul Cha
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kee-Joon Lee
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kwang-Mahn Kim
- Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jin-Man Kim
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Dohyun Kim
- Department of Conservative Dentistry, Oral Science Research Center, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jae-Sung Kwon
- BK21 FOUR Project, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Corresponding author. Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Corresponding author. Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Sung-Hwan Choi
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Corresponding author. Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
43
|
Zhang Y, Chen H, Li J. Recent advances on gelatin methacrylate hydrogels with controlled microstructures for tissue engineering. Int J Biol Macromol 2022; 221:91-107. [DOI: 10.1016/j.ijbiomac.2022.08.171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 12/12/2022]
|
44
|
Barnett H, Shevchuk M, Peppas NA, Caldorera-Moore M. Influence of extracellular cues of hydrogel biomaterials on stem cell fate. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1324-1347. [PMID: 35297325 DOI: 10.1080/09205063.2022.2054398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
Tissue engineering is a multidisciplinary field that focuses on creating functional tissue through the combination of biomimetic scaffolds, a cell source, and biochemical/physiochemical cues. Stem cells are often used as the cell source due to their multipotent properties and autologous sourcing; however, the combination of physical and chemical cues that regulate their behavior creates challenges in reproducibly directing them to a specific fate. Hydrogel biomaterials are widely explored as tissue scaffolds due to their innate biomimetic properties and tailorability. For these constructs to be successful, properties such as surface chemistry and spatial configuration, stiffness, and degradability of the biomaterial used for the scaffold framework should be analogous to the natural environment of the tissue they are repairing/replacing. This is imperative, as cues from the surrounding extracellular matrix (ECM) influence stem cell behavior and direct cell differentiation to a specific lineage. Hydrogels offer great promise as tools to control stem cell fate, as researchers can modulate the degradation rates, mechanical properties, swelling behavior, and chemical properties of the biomaterial scaffold to mimic the instructive cues of the native ECM. Discussion of the advantages and challenges of utilizing hydrogel biomaterials as the basis of tissue scaffolds is reviewed herein, as well as specific examples of hydrogels in tissue engineering and advances in hydrogel research to achieve desired cell phenotypes.
Collapse
Affiliation(s)
- Haley Barnett
- School of Sciences, University of Louisiana Monroe, Monroe, LA, USA
| | - Mariya Shevchuk
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute of Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, and Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Mary Caldorera-Moore
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA, USA
| |
Collapse
|
45
|
Hu X, Zheng J, Hu Q, Liang L, Yang D, Cheng Y, Li SS, Chen LJ, Yang Y. Smart acoustic 3D cell construct assembly with high-resolution. Biofabrication 2022; 14. [PMID: 35764072 DOI: 10.1088/1758-5090/ac7c90] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/22/2022] [Indexed: 11/12/2022]
Abstract
Precise and flexible three-dimensional (3D) cell construct assembly using external forces or fields can produce micro-scale cellular architectures with intercellular connections, which is an important prerequisite to reproducing the structures and functions of biological systems. Currently, it is also a substantial challenge in the bioengineering field. Here, we propose a smart acoustic 3D cell assembly strategy that utilizes a 3D printed module and hydrogel sheets. Digitally controlled six wave beams offer a high degree of freedom (including wave vector combination, frequency, phase, and amplitude) that enables versatile biomimetic micro cellular patterns in hydrogel sheets. Further, replaceable frames can be used to fix the acoustic-built micro-scale cellular structures in these sheets, enabling user-defined hierarchical or heterogeneous constructs through layer-by-layer assembly. This strategy can be employed to construct vasculature with different diameters and lengths, composed of human umbilical vein endothelial cells and smooth muscle cells. These constructs can also induce controllable vascular network formation. Overall, the findings of this work extend the capabilities of acoustic cell assembly into 3D space, offering advantages including innovative, flexible, and precise patterning, and displaying great potential for the manufacture of various artificial tissue structures that duplicate in vivo functions.
Collapse
Affiliation(s)
- Xuejia Hu
- School of Electronic Science and Engineering, Xiamen University, Xiamen University, No. 422 Siming south road, Xiamen, Fujian, 361005, CHINA
| | - Jingjing Zheng
- School of physics and engineering, Wuhan University, luojia mountain street, Wuhan, Wuhan, Hubei, 430072, CHINA
| | - Qinghao Hu
- School of physics and engineering, Wuhan University, luojia street, Wuhan, Wuhan, Hubei, 430072, CHINA
| | - Li Liang
- School of Physics and Electronic Technology, Anhui Normal University, No. 189 of jiuhua south road, Wuhu, Wuhu, Anhui, 241000, CHINA
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, No. 238, Jiefang road, Wuhan, Hubei, 430060, CHINA
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, No. 238, Jiefang road, Wuhan, Hubei, 430060, CHINA
| | - Sen-Sen Li
- School of Electronic Science and Engineering, Xiamen University, Xiamen University, No. 422 Siming south road, Xiamen, Fujian, 361005, CHINA
| | - Lu-Jian Chen
- School of Electronic Science and Engineering, Xiamen University, Xiamen University, No. 422 Siming south road, Xiamen, Fujian, 361005, CHINA
| | - Yi Yang
- School of physics and engineering, Wuhan University, luojia street, Wuhan, Wuhan, Hubei, 430072, CHINA
| |
Collapse
|
46
|
Abstract
A novel composite hydrogel was developed that shows remarkable similarities to load bearing biological tissues. The composite gel consisting of a poly(vinyl alcohol (PVA) matrix filled with poly(acrylic acid) (PAA) microgel particles exhibits osmotic and mechanical properties that are qualitatively different from regular gels. In the PVA/PAA system the swollen PAA particles "inflate" the PVA network. The swelling of the PAA is limited by the tensile stress Pel developing in the PVA matrix. Pel increases with increasing swelling degree, which is opposite to the decrease of the elastic pressure observed in regular gels. The maximum tensile stress Pmaxel can be identified as a quantity that defines the load bearing ability of the composite gel. Systematic osmotic swelling pressure measurements have been made on PVA/PAA gels to determine the effects of PVA stiffness, PAA crosslink density, and Ca2+ ion concentration on Pmaxel. It is found that Pmaxel increases with the stiffness of the PVA matrix, and decreases with (i) increasing crosslink density of the PAA and (ii) increasing Ca2+ ion concentration. Small angle neutron scattering (SANS) measurements indicate only a weak interaction between the PVA and PAA gels. It is demonstrated that the osmotic swelling pressure of PVA/PAA composite gels reproduces the osmotic behavior of healthy and osteoarthritic cartilage.
Collapse
Affiliation(s)
- Ferenc Horkay
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Peter J Basser
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Ashammakhi N, GhavamiNejad A, Tutar R, Fricker A, Roy I, Chatzistavrou X, Hoque Apu E, Nguyen KL, Ahsan T, Pountos I, Caterson EJ. Highlights on Advancing Frontiers in Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:633-664. [PMID: 34210148 PMCID: PMC9242713 DOI: 10.1089/ten.teb.2021.0012] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/15/2021] [Indexed: 01/05/2023]
Abstract
The field of tissue engineering continues to advance, sometimes in exponential leaps forward, but also sometimes at a rate that does not fulfill the promise that the field imagined a few decades ago. This review is in part a catalog of success in an effort to inform the process of innovation. Tissue engineering has recruited new technologies and developed new methods for engineering tissue constructs that can be used to mitigate or model disease states for study. Key to this antecedent statement is that the scientific effort must be anchored in the needs of a disease state and be working toward a functional product in regenerative medicine. It is this focus on the wildly important ideas coupled with partnered research efforts within both academia and industry that have shown most translational potential. The field continues to thrive and among the most important recent developments are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies that warrant special attention. Developments in the aforementioned areas as well as future directions are highlighted in this article. Although several early efforts have not come to fruition, there are good examples of commercial profitability that merit continued investment in tissue engineering. Impact statement Tissue engineering led to the development of new methods for regenerative medicine and disease models. Among the most important recent developments in tissue engineering are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies. These technologies and an understanding of them will have impact on the success of tissue engineering and its translation to regenerative medicine. Continued investment in tissue engineering will yield products and therapeutics, with both commercial importance and simultaneous disease mitigation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, Michigan, USA
| | - Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Annabelle Fricker
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Ehsanul Hoque Apu
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Taby Ahsan
- RoosterBio, Inc., Frederick, Maryland, USA
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
| | - Edward J. Caterson
- Division of Plastic Surgery, Department of Surgery, Nemours/Alfred I. du Pont Hospital for Children, Wilmington, Delaware, USA
| |
Collapse
|
48
|
Astor TL, Borenstein JT. The microfluidic artificial lung: Mimicking nature's blood path design to solve the biocompatibility paradox. Artif Organs 2022; 46:1227-1239. [PMID: 35514275 DOI: 10.1111/aor.14266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/28/2022]
Abstract
The increasing prevalence of chronic lung disease worldwide, combined with the emergence of multiple pandemics arising from respiratory viruses over the past century, highlights the need for safer and efficacious means for providing artificial lung support. Mechanical ventilation is currently used for the vast majority of patients suffering from acute and chronic lung failure, but risks further injury or infection to the patient's already compromised lung function. Extracorporeal membrane oxygenation (ECMO) has emerged as a means of providing direct gas exchange with the blood, but limited access to the technology and the complexity of the blood circuit have prevented the broader expansion of its use. A promising avenue toward simplifying and minimizing complications arising from the blood circuit, microfluidics-based artificial organ support, has emerged over the past decade as an opportunity to overcome many of the fundamental limitations of the current standard for ECMO cartridges, hollow fiber membrane oxygenators. The power of microfluidics technology for this application stems from its ability to recapitulate key aspects of physiological microcirculation, including the small dimensions of blood vessel structures and gas transfer membranes. An even greater advantage of microfluidics, the ability to configure blood flow patterns that mimic the smooth, branching nature of vascular networks, holds the potential to reduce the incidence of clotting and bleeding and to minimize reliance on anticoagulants. Here, we summarize recent progress and address future directions and goals for this potentially transformative approach to artificial lung support.
Collapse
Affiliation(s)
- Todd L Astor
- Biomembretics, Inc., Boston, Massachusetts, USA.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
49
|
Yang D, Xie L, Mao X, Gong L, Peng X, Peng Q, Wang T, Liu Q, Zeng H, Zhang H. Probing Hydrophobic Interactions between Polymer Surfaces and Air Bubbles or Oil Droplets: Effects of Molecular Weight and Surfactants. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:5257-5268. [PMID: 34787428 DOI: 10.1021/acs.langmuir.1c02635] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Hydrophobic interaction plays an important role in numerous interfacial phenomena and biophysical and industrial processes. In this work, polystyrene (PS) was used as a model hydrophobic polymer for investigating its hydrophobic interaction with highly deformable objects (i.e., air bubbles and oil droplets) in aqueous solutions. The effects of polymer molecular weight, solvent (i.e., addition of ethanol to water), the presence of surface-active species, and hydrodynamic conditions were investigated, via direct surface force measurements using the bubble/drop probe atomic force microscopy (AFM) technique and theoretical calculations based on the Reynolds lubrication theory and augmented Young-Laplace equation by including the effect of disjoining pressure. It was found that the PS of low molecular weight (i.e., PS590 and PS810) showed slightly weaker hydrophobic interactions with air bubbles or oil droplets, as compared to glassy PS of higher molecular weight (i.e., PS1110, PS2330, PS46300, and PS1M). The hydrophobic interaction between PS and air bubbles in a 1 M NaCl aqueous solution with 10 vol % ethanol was weaker than that in the bare aqueous solution. Such effects on the hydrophobic interactions are possibly achieved by influencing the structuring/ordering of water molecules close to the hydrophobic polymer surfaces by tuning the surface chain mobility and surface roughness of polymers. It was found that the addition of three surface-active species, i.e., cetyltrimethylammonium chloride (CTAC), Pluronic F-127, and sodium dodecyl sulfate (SDS), to the aqueous media could suppress the attachment of the hydrophobic polymer and air bubbles or oil droplets, most likely caused by the additional steric repulsion due to the adsorbed surface-active species at the bubble/polymer/oil interfaces. Our results have improved the fundamental understanding of the interaction mechanisms between hydrophobic polymers and gas bubbles or oil droplets, with useful implications on developing effective methods for modulating the related interfacial interactions in many engineering applications.
Collapse
Affiliation(s)
- Diling Yang
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Lei Xie
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Xiaohui Mao
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Lu Gong
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Xuwen Peng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Qiongyao Peng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Tao Wang
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Qi Liu
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Hongbo Zeng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Hao Zhang
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| |
Collapse
|
50
|
Lv Q, Sun X, Ye L, Liang H. Stiff and strong hydrogel tube with great mechanical properties and high stability in various solutions. J Mater Chem B 2022; 10:3126-3137. [PMID: 35348565 DOI: 10.1039/d2tb00124a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hydrogel tubes are widely used in fields such as artificial blood vessels, drug delivery, biomedical scaffolds and cell adhesion, yet their application is often limited by unsatisfactory mechanical properties and poor stability in various solutions. Herein, a novel hydrogel tube exhibiting a remarkable mechanical performance and stability in various solutions is prepared by introducing a dual physically cross-linked double network (DN) hydrogel matrix. The obtained hydrogel tube can withstand ∼60 N load without fracture and be stretched to over twice its original length before and after immersing in various solutions. The great mechanical properties and stability in various solutions of hydrogel tubes are due to the introduction of a dual physically cross-linked poly(acrylamide-co-acrylic acid)/carboxymethylcellulose sodium/Fe3+ DN hydrogel, which possesses high elastic modulus (3.71 MPa), fracture energy (15.4 kJ m-2), and great stability in various solutions. In addition, the hydrogel tubes with different thickness, diameters, shapes and the multiple branched hydrogel tubes can also be fabricated to enable further functionalization for application requirements. Therefore, this new type of hydrogel tube presents tremendous potential for applications in biomedical and engineering fields.
Collapse
Affiliation(s)
- Qiong Lv
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Xingyue Sun
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Lina Ye
- School of Material Science and Engineering, Anhui University, Hefei, Anhui 230601, China.
| | - Haiyi Liang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230026, China. .,IAT-Chungu Joint Laboratory for Additive Manufacturing, Anhui Chungu 3D Printing Institute of Intelligent Equipment and Industrial Technology, Wuhu, Anhui 241200, China
| |
Collapse
|