1
|
Marjan T, Lafuente-Gómez N, Rampal A, Mooney DJ, Peyton SR, Qazi TH. Cell-Instructive Biomaterials with Native-Like Biochemical Complexity. Annu Rev Biomed Eng 2025; 27:185-209. [PMID: 39874600 PMCID: PMC12045723 DOI: 10.1146/annurev-bioeng-120823-020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Biochemical signals in native tissue microenvironments instruct cell behavior during many biological processes ranging from developmental morphogenesis and tissue regeneration to tumor metastasis and disease progression. The detection and characterization of these signals using spatial and highly resolved quantitative methods have revealed their existence as matricellular proteins in the matrisome, some of which are bound to the extracellular matrix while others are freely diffusing. Including these biochemical signals in engineered biomaterials can impart enhanced functionality and native-like complexity, ultimately benefiting efforts to understand, model, and treat various diseases. In this review, we discuss advances in characterizing, mimicking, and harnessing biochemical signals in developing advanced engineered biomaterials. An overview of the diverse forms in which these biochemical signals exist and their effects on intracellular signal transduction is also provided. Finally, we highlight the application of biochemically complex biomaterials in the three broadly defined areas of tissue regeneration, immunoengineering, and organoid morphogenesis.
Collapse
Affiliation(s)
- Tuba Marjan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Nuria Lafuente-Gómez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Akaansha Rampal
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
2
|
Moghaddam AS, Dunne K, Breyer W, Wu Y, Pashuck ET. Hydrogels with multiple RGD presentations increase cell adhesion and spreading. Acta Biomater 2025:S1742-7061(25)00288-0. [PMID: 40254231 DOI: 10.1016/j.actbio.2025.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
A key challenge in designing hydrogels for cell culture is replicating the cell-matrix interactions found in tissues. Cells use integrins to bind their local matrix and form adhesions in which integrins dynamically move on the cell membrane while applying significant forces to the local matrix. Identifying the important biomaterial features for these interactions is challenging because it is difficult to independently adjust variables such as matrix stiffness, stress relaxation, the mobility of adhesion ligands, and the ability of these ligands to support cellular forces. In this work, we designed a hydrogel platform consisting of interpenetrating polymer networks of covalently crosslinked poly(ethylene glycol) (PEG) and self-assembled peptide amphiphiles (PA). We can tune the viscoelasticity of the hydrogel by modulating the composition of both networks. Ligand mobility can be adjusted independently of the matrix mechanical properties by attaching the arginine-glycine-aspartic acid (RGD) cell adhesion ligand to either the covalent PEG network, the dynamic PA network, or both networks at once. We find that endothelial cell adhesion formation and spreading is maximized in soft gels in which adhesion ligands are present on both the covalent and non-covalent networks. The dynamic nature of adhesion domains, coupled with their ability to exert substantial forces on the matrix, suggests that having different presentations of RGD ligands which are either mobile or capable of withstanding significant forces is needed to mimic different aspects of complex cell-matrix adhesions. These results will contribute to the design of hydrogels that better recapitulate physiological cell-matrix interactions. STATEMENT OF SIGNIFICANCE: Creating artificial environments that accurately mimic how cells interact with their surrounding matrix in natural tissues remains a fundamental challenge in biomaterials science. This study introduces a dual-network hydrogel platform that independently controls mechanical properties and adhesion ligand mobility by combining stable and dynamic polymer networks. A significant body of work has shown that matrix viscoelasticity and adhesion ligand mobility are important for cell adhesion and spreading. Our work builds on this by showing that endothelial cells function optimally when they can simultaneously engage with both mobile adhesion sites and force-resistant anchoring points, independent of matrix viscoelasticity. These insights will guide the design of more physiologically relevant hydrogels for tissue engineering applications and disease modeling.
Collapse
Affiliation(s)
| | | | - Wendy Breyer
- Department of Chemistry, Lehigh University, Bethlehem, PA 18015, USA
| | | | | |
Collapse
|
3
|
Ge B, Xie Q, Wu D, Xu J, Jiao H, Zhao D, Li J. Hydrogels as drug delivery platforms for orthopedic diseases treatment: A review. Int J Biol Macromol 2025; 304:140902. [PMID: 39947563 DOI: 10.1016/j.ijbiomac.2025.140902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/20/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
The skeletal system serves as a crucial support structure for the human body, any damage or disease to bones can result in prolonged pain, impaired mobility, and other negative outcomes. For the treatment of bone diseases, with the in-depth study of the therapeutic mechanism, various small molecule drugs, cells, cytokines, growth factors, bioactive ions, collectively referred to as "drugs" in this context, are increasingly investigated for their potential application in surgical procedures, defect repair, or treatment of diseased bone regions. However, various challenges, including, low stability, the necessity for precise dosage control, are encountered in the administration of drugs. Consequently, the advancement of drug delivery platforms is crucial to safeguard drug efficacy and address the requirement for dosage regulation. Given the attributes of current drug delivery platforms, hydrogels exhibit favorable biocompatibility and offer the ability to easily regulate drug loading and release. As a carrier with diverse properties, abundant varieties, optimal performance, hydrogels present a promising solution in drug delivery. This paper aims to analyze the potential of hydrogel as a delivery platform for treating orthopedics diseases by reviewing the characteristics of hydrogel delivery systems, mechanisms of drug binding, current research findings, and projecting future developments in this field.
Collapse
Affiliation(s)
- Bing Ge
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Qinwen Xie
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Di Wu
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Jianfeng Xu
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Haolin Jiao
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Dewei Zhao
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China.
| | - Junlei Li
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China.
| |
Collapse
|
4
|
Cota Quintero JL, Ramos-Payán R, Romero-Quintana JG, Ayala-Ham A, Bermúdez M, Aguilar-Medina EM. Hydrogel-Based Scaffolds: Advancing Bone Regeneration Through Tissue Engineering. Gels 2025; 11:175. [PMID: 40136878 PMCID: PMC11942283 DOI: 10.3390/gels11030175] [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: 01/29/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Bone tissue engineering has emerged as a promising approach to addressing the limitations of traditional bone grafts for repairing bone defects. This regenerative medicine strategy leverages biomaterials, growth factors, and cells to create a favorable environment for bone regeneration, mimicking the body's natural healing process. Among the various biomaterials explored, hydrogels (HGs), a class of three-dimensional, hydrophilic polymer networks, have gained significant attention as scaffolds for bone tissue engineering. Thus, this review aimed to investigate the potential of natural and synthetic HGs, and the molecules used for its functionalization, for enhanced bone tissue engineering applications. HGs offer several advantages such as scaffolds, including biocompatibility, biodegradability, tunable mechanical properties, and the ability to encapsulate and deliver bioactive molecules. These properties make them ideal candidates for supporting cell attachment, proliferation, and differentiation, ultimately guiding the formation of new bone tissue. The design and optimization of HG-based scaffolds involve adapting their composition, structure, and mechanical properties to meet the specific requirements of bone regeneration. Current research focuses on incorporating bioactive molecules, such as growth factors and cytokines, into HG scaffolds to further enhance their osteoinductive and osteoconductive properties. Additionally, strategies to improve the mechanical strength and degradation kinetics of HGs are being explored to ensure long-term stability and support for new bone formation. The development of advanced HG-based scaffolds holds great potential for revolutionizing bone tissue engineering and providing effective treatment options for patients with bone defects.
Collapse
Affiliation(s)
- Juan Luis Cota Quintero
- Faculty of Biology, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico;
| | - Rosalío Ramos-Payán
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico; (R.R.-P.); (J.G.R.-Q.)
| | - José Geovanni Romero-Quintana
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico; (R.R.-P.); (J.G.R.-Q.)
| | - Alfredo Ayala-Ham
- Faculty of Odontology, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico;
| | - Mercedes Bermúdez
- Faculty of Odontology, Autonomous University of Chihuahua, Circuito Universitario Campus I, Chihuahua 31000, Chihuahua, Mexico;
| | - Elsa Maribel Aguilar-Medina
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Josefa Ortiz de Domínguez s/n y Avenida de las Américas, Culiacan 80010, Sinaloa, Mexico; (R.R.-P.); (J.G.R.-Q.)
| |
Collapse
|
5
|
Thompson GB, Lee J, Kamani KM, Flores-Velasco N, Rogers SA, Harley BAC. Granular hydrogels as brittle yield stress fluids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.639638. [PMID: 40060491 PMCID: PMC11888328 DOI: 10.1101/2025.02.22.639638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
While granular hydrogels are increasingly used in biomedical applications, methods to capture their rheological behavior generally consider shear-thinning and self-healing properties or produce ensemble metrics such as the dynamic moduli. Analytical approaches paired with common oscillatory shear tests can describe not only solid-like and fluid-like behavior of granular hydrogels but also transient characteristics inherent in yielding and unyielding processes. Combining oscillatory shear testing with consideration of Brittility (Bt) via the Kamani-Donley-Rogers (KDR) model, we show granular hydrogels behave as brittle yield stress fluids with complex transient rheology. We quantify steady and transient rheology as a function of microgel (composition; diameter) and granular (packing; droplet heterogeneity) assembly properties for mixtures of polyethylene glycol and gelatin microgels. The KDR model with Bt captures granular hydrogel behavior for a wide range of design parameters, reducing the complex transient rheology to a determination of model parameters. We describe the impact of composition on rheological behavior and model parameters in monolithic and mixed granular hydrogels. The model robustly captures self-healing behavior and reveals granular relaxation time depends on strain amplitude. This quantitative framework is an important step toward rational design of granular hydrogels for applications ranging from injection and in situ stabilization to 3D bioprinting.
Collapse
Affiliation(s)
- G B Thompson
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - J Lee
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - K M Kamani
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - N Flores-Velasco
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - S A Rogers
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - B A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
6
|
Rozans SJ, Wu Y, Moghaddam AS, Pashuck ET. A Streamlined High-Throughput LC-MS Assay for Quantifying Peptide Degradation in Cell Culture. J Biomed Mater Res A 2025; 113:e37864. [PMID: 39806927 PMCID: PMC11913071 DOI: 10.1002/jbm.a.37864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
Peptides are widely used in biomaterials due to their ease of synthesis, ability to signal cells, and modify the properties of biomaterials. A key benefit of using peptides is that they are natural substrates for cell-secreted enzymes, which creates the possibility of utilizing cell-secreted enzymes for tuning cell-material interactions. However, these enzymes can also induce unwanted degradation of bioactive peptides in biomaterials, or in peptide therapies. Liquid chromatography-mass spectrometry (LC-MS) is a widely used, powerful methodology that can separate complex mixtures of molecules and quantify numerous analytes within a single run. There are several challenges in using LC-MS for the multiplexed quantification of cell-induced peptide degradation, including the need for nondegradable internal standards and the identification of optimal sample storage conditions. Another problem is that cell culture media and biological samples typically contain both proteins and lipids that can accumulate on chromatography columns and degrade their performance. Removing these constituents can be expensive, time-consuming, and increases sample variability. However, loading unpurified samples onto the column without removing lipids and proteins will foul the column. Here, we show that directly injecting complex, unpurified samples onto the LC-MS without any purification enables rapid and accurate quantification of peptide concentration and that hundreds of LC-MS runs can be done on a single column without significantly diminishing the ability to quantify the degradation of peptide libraries. To understand how repeated injections degrade column performance, a model library was injected into the LC-MS hundreds of times. It was then determined that column failure is evident when hydrophilic peptides are no longer retained on the column and that failure can be easily identified by using standard peptide mixtures for column benchmarking. In total, this work introduces a simple and effective method for simultaneously quantifying the degradation of dozens of peptides in cell culture. By providing a streamlined and cost-effective method for the direct quantification of peptide degradation in complex biological samples, this work enables more efficient assessment of peptide stability and functionality, facilitating the development of advanced biomaterials and peptide-based therapies.
Collapse
Affiliation(s)
- Samuel J Rozans
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | | | - E Thomas Pashuck
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| |
Collapse
|
7
|
Thompson GB, Gilchrist AE, Lam VM, Nunes AC, Payan BA, Mora-Boza A, Serrano JF, García AJ, Harley BAC. Gelatin maleimide microgels for hematopoietic progenitor cell encapsulation. J Biomed Mater Res A 2024; 112:2124-2135. [PMID: 38894666 PMCID: PMC11464195 DOI: 10.1002/jbm.a.37765] [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: 02/02/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/21/2024]
Abstract
Hematopoietic stem cells (HSCs) are the apical cells of the hematopoietic system, giving rise to cells of the blood and lymph lineages. HSCs reside primarily within bone marrow niches that contain matrix and cell-derived signals that help inform stem cell fate. Aspects of the bone marrow microenvironment have been captured in vitro by encapsulating cells within hydrogel matrices that mimic native mechanical and biochemical properties. Hydrogel microparticles, or microgels, are increasingly being used to assemble granular biomaterials for cell culture and noninvasive delivery applications. Here, we report the optimization of a gelatin maleimide hydrogel system to create monodisperse gelatin microgels via a flow-focusing microfluidic process. We report characteristic hydrogel stiffness, stability, and swelling characteristics as well as encapsulation of murine hematopoietic stem and progenitor cells, and mesenchymal stem cells within microgels. Microgels support cell viability, confirming compatibility of the microfluidic encapsulation process with these sensitive bone marrow cell populations. Overall, this work presents a microgel-based gelatin maleimide hydrogel as a foundation for future development of a multicellular artificial bone marrow culture system.
Collapse
Affiliation(s)
- Gunnar B Thompson
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis, USA
| | - Vincent M Lam
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Alison C Nunes
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Brittany A Payan
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Ana Mora-Boza
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Julio F Serrano
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- George Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
8
|
Bai L, Zhou D, Li G, Liu J, Chen X, Su J. Engineering bone/cartilage organoids: strategy, progress, and application. Bone Res 2024; 12:66. [PMID: 39567500 PMCID: PMC11579019 DOI: 10.1038/s41413-024-00376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
The concept and development of bone/cartilage organoids are rapidly gaining momentum, providing opportunities for both fundamental and translational research in bone biology. Bone/cartilage organoids, essentially miniature bone/cartilage tissues grown in vitro, enable the study of complex cellular interactions, biological processes, and disease pathology in a representative and controlled environment. This review provides a comprehensive and up-to-date overview of the field, focusing on the strategies for bone/cartilage organoid construction strategies, progresses in the research, and potential applications. We delve into the significance of selecting appropriate cells, matrix gels, cytokines/inducers, and construction techniques. Moreover, we explore the role of bone/cartilage organoids in advancing our understanding of bone/cartilage reconstruction, disease modeling, drug screening, disease prevention, and treatment strategies. While acknowledging the potential of these organoids, we discuss the inherent challenges and limitations in the field and propose potential solutions, including the use of bioprinting for organoid induction, AI for improved screening processes, and the exploration of assembloids for more complex, multicellular bone/cartilage organoids models. We believe that with continuous refinement and standardization, bone/cartilage organoids can profoundly impact patient-specific therapeutic interventions and lead the way in regenerative medicine.
Collapse
Affiliation(s)
- Long Bai
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, China
| | - Dongyang Zhou
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Guangfeng Li
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Jinlong Liu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| |
Collapse
|
9
|
Rozans SJ, Moghaddam AS, Pashuck ET. A Streamlined High-Throughput LC-MS Assay for Quantifying Peptide Degradation in Cell Culture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617883. [PMID: 39463983 PMCID: PMC11507709 DOI: 10.1101/2024.10.11.617883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Peptides are widely used in biomaterials due to their easy of synthesis, ability to signal cells, and modify the properties of biomaterials. A key benefit of using peptides is that they are natural substrates for cell-secreted enzymes, which creates the possibility of utilizing cell-secreted enzymes for tuning cell-material interactions. However, these enzymes can also induce unwanted degradation of bioactive peptides in biomaterials, or in peptide therapies. Liquid chromatography-mass spectrometry (LC-MS) is a widely used, powerful methodology that can separate complex mixtures of molecules and quantify numerous analytes within a single run. There are several challenges in using LC-MS for the multiplexed quantification of cell-induced peptide degradation, including the need for non-degradable internal standards and the identification of optimal sample storage conditions. Another problem is that cell culture media and biological samples typically contain both proteins and lipids that can accumulate on chromatography columns and degrade their performance. However, removing these constituents can be expensive, time consuming, and increases sample variability. Here we show that directly injecting samples onto the LC-MS without any purification enables rapid and accurate quantification of peptide concentration, and that hundreds of LC-MS runs can be done on a single column without a significantly diminish the ability to quantify the degradation of peptide libraries. We also show that column failure is evident when hydrophilic peptides fail to be retained on the column, and this can be easily identified using standard peptide mixtures for column benchmarking. In total, this work introduces a simple and effective method for simultaneously quantifying the degradation of dozens of peptides in cell culture. By providing a streamlined and cost-effective method for the direct quantification of peptide degradation in complex biological samples, this work enables more efficient assessment of peptide stability and functionality, facilitating the development of advanced biomaterials and peptide-based therapies.
Collapse
Affiliation(s)
- Samuel J Rozans
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA, 18015
| | | | - E Thomas Pashuck
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA, 18015
| |
Collapse
|
10
|
Baldassarri I, Tavakol DN, Graney PL, Chramiec AG, Hibshoosh H, Vunjak-Novakovic G. An engineered model of metastatic colonization of human bone marrow reveals breast cancer cell remodeling of the hematopoietic niche. Proc Natl Acad Sci U S A 2024; 121:e2405257121. [PMID: 39374382 PMCID: PMC11494322 DOI: 10.1073/pnas.2405257121] [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: 03/19/2024] [Accepted: 08/16/2024] [Indexed: 10/09/2024] Open
Abstract
Incomplete understanding of metastatic disease mechanisms continues to hinder effective treatment of cancer. Despite remarkable advancements toward the identification of druggable targets, treatment options for patients in remission following primary tumor resection remain limited. Bioengineered human tissue models of metastatic sites capable of recreating the physiologically relevant milieu of metastatic colonization may strengthen our grasp of cancer progression and contribute to the development of effective therapeutic strategies. We report the use of an engineered tissue model of human bone marrow (eBM) to identify microenvironmental cues regulating cancer cell proliferation and to investigate how triple-negative breast cancer (TNBC) cell lines influence hematopoiesis. Notably, individual stromal components of the bone marrow niche (osteoblasts, endothelial cells, and mesenchymal stem/stromal cells) were each critical for regulating tumor cell quiescence and proliferation in the three-dimensional eBM niche. We found that hematopoietic stem and progenitor cells (HSPCs) impacted TNBC cell growth and responded to cancer cell presence with a shift of HSPCs (CD34+CD38-) to downstream myeloid lineages (CD11b+CD14+). To account for tumor heterogeneity and show proof-of-concept ability for patient-specific studies, we demonstrate that patient-derived tumor organoids survive and proliferate in the eBM, resulting in distinct shifts in myelopoiesis that are similar to those observed for aggressively metastatic cell lines. We envision that this human tissue model will facilitate studies of niche-specific metastatic progression and individualized responses to treatment.
Collapse
Affiliation(s)
- Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Pamela L. Graney
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Alan G. Chramiec
- Department of Biomedical Engineering, Columbia University, New York, NY10025
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University, New York, NY10032
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Department of Medicine, Columbia University, New York, NY10032
- College of Dental Medicine, Columbia University, New York, NY10032
| |
Collapse
|
11
|
Moghaddam AS, Dunne K, Breyer W, Wu Y, Pashuck ET. Hydrogels with Independently Controlled Adhesion Ligand Mobility and Viscoelasticity Increase Cell Adhesion and Spreading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614501. [PMID: 39386463 PMCID: PMC11463488 DOI: 10.1101/2024.09.23.614501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
A primary objective in designing hydrogels for cell culture is recreating the cell-matrix interactions found within human tissues. Identifying the most important biomaterial features for these interactions is challenging because it is difficult to independently adjust variables such as matrix stiffness, stress relaxation, the mobility of adhesion ligands and the ability of these ligands to support cellular forces. In this work we designed a hydrogel platform consisting of interpenetrating polymer networks of covalently crosslinked poly(ethylene glycol) (PEG) and self-assembled peptide amphiphiles (PA). We can tailor the storage modulus of the hydrogel by altering the concentration and composition of each network, and we can tune the stress relaxation half-life through the non-covalent bonding in the PA network. Ligand mobility can be adjusted independently of the matrix mechanical properties by attaching the RGD cell adhesion ligand to either the covalent PEG network, the dynamic PA network, or both networks at once. Interestingly, our findings show that endothelial cell adhesion formation and spreading is maximized in soft, viscoelastic gels in which RGD adhesion ligands are present on both the covalent PEG and non-covalent PA networks. The dynamic nature of cell adhesion domains, coupled with their ability to exert substantial forces on the matrix, suggests that having different presentations of RGD ligands which are either mobile or are capable of withstanding significant forces are needed mimic different aspects of complex cell-matrix adhesions. By demonstrating how different presentations of RGD ligands affect cell behavior independently of viscoelastic properties, these results contribute to the rational design of hydrogels that facilitate desired cell-matrix interactions, with the potential of improving in vitro models and regenerative therapies.
Collapse
Affiliation(s)
| | - Katelyn Dunne
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| | - Wendy Breyer
- Department of Chemistry, Lehigh University, Bethlehem PA, USA, 18015
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| | - E Thomas Pashuck
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| |
Collapse
|
12
|
Nuckhir M, Withey D, Cabral S, Harrison H, Clarke RB. State of the Art Modelling of the Breast Cancer Metastatic Microenvironment: Where Are We? J Mammary Gland Biol Neoplasia 2024; 29:14. [PMID: 39012440 PMCID: PMC11252219 DOI: 10.1007/s10911-024-09567-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/09/2024] [Indexed: 07/17/2024] Open
Abstract
Metastatic spread of tumour cells to tissues and organs around the body is the most frequent cause of death from breast cancer. This has been modelled mainly using mouse models such as syngeneic mammary cancer or human in mouse xenograft models. These have limitations for modelling human disease progression and cannot easily be used for investigation of drug resistance and novel therapy screening. To complement these approaches, advances are being made in ex vivo and 3D in vitro models, which are becoming progressively better at reliably replicating the tumour microenvironment and will in the future facilitate drug development and screening. These approaches include microfluidics, organ-on-a-chip and use of advanced biomaterials. The relevant tissues to be modelled include those that are frequent and clinically important sites of metastasis such as bone, lung, brain, liver for invasive ductal carcinomas and a distinct set of common metastatic sites for lobular breast cancer. These sites all have challenges to model due to their unique cellular compositions, structure and complexity. The models, particularly in vivo, provide key information on the intricate interactions between cancer cells and the native tissue, and will guide us in producing specific therapies that are helpful in different context of metastasis.
Collapse
Affiliation(s)
- Mia Nuckhir
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - David Withey
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - Sara Cabral
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK
| | - Hannah Harrison
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK.
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Oglesby Cancer Research Building, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M20 4GJ, UK.
| |
Collapse
|
13
|
Richbourg NR, Irakoze N, Kim H, Peyton SR. Outlook and opportunities for engineered environments of breast cancer dormancy. SCIENCE ADVANCES 2024; 10:eadl0165. [PMID: 38457510 PMCID: PMC10923521 DOI: 10.1126/sciadv.adl0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/01/2024] [Indexed: 03/10/2024]
Abstract
Dormant, disseminated breast cancer cells resist treatment and may relapse into malignant metastases after decades of quiescence. Identifying how and why these dormant breast cancer cells are triggered into outgrowth is a key unsolved step in treating latent, metastatic breast cancer. However, our understanding of breast cancer dormancy in vivo is limited by technical challenges and ethical concerns with triggering the activation of dormant breast cancer. In vitro models avoid many of these challenges by simulating breast cancer dormancy and activation in well-controlled, bench-top conditions, creating opportunities for fundamental insights into breast cancer biology that complement what can be achieved through animal and clinical studies. In this review, we address clinical and preclinical approaches to treating breast cancer dormancy, how precisely controlled artificial environments reveal key interactions that regulate breast cancer dormancy, and how future generations of biomaterials could answer further questions about breast cancer dormancy.
Collapse
Affiliation(s)
- Nathan R. Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst Amherst, MA 01003, USA
| |
Collapse
|
14
|
Gong L, Zhu J, Yang Y, Qiao S, Ma L, Wang H, Zhang Y. Effect of polyethylene glycol on polysaccharides: From molecular modification, composite matrixes, synergetic properties to embeddable application in food fields. Carbohydr Polym 2024; 327:121647. [PMID: 38171672 DOI: 10.1016/j.carbpol.2023.121647] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024]
Abstract
Polyethylene glycol (PEG) is a flexible, water-soluble, non-immunogenic, as well as biocompatible polymer, and it could synergize with polysaccharides for food applications. The molecular modification strategies, including covalent bond interactions (amino groups, carboxyl groups, aldehyde groups, tosylate groups, etc.), and non-covalent bond interactions (hydrogen bonding, electrostatic interactions, etc.) on PEG molecular chains are discussed. Its versatile structure, group modifiability, and amphiphilic block buildability could improve the functions of polysaccharides (e.g., chitosan, cellulose, starch, alginate, etc.) and adjust the properties of combined PEG/polysaccharides with outstanding chain tunability and matrix processability owing to plasticizing effects, compatibilizing effects, steric stabilizing effects and excluded volume effects by PEG, for achieving the diverse performance targets. The synergetic properties of PEG/polysaccharides with remarkable architecture were summarized, including mechanical properties, antibacterial activity, antioxidant performance, self-healing properties, carrier and delivery characteristics. The PEG/polysaccharides with excellent combined properties and embeddable merits illustrate potential applications including food packaging, food intelligent indication/detection, food 3D printing and nutraceutical food absorption. Additionally, prospects (like food innovation and preferable nutrient utilization) and key challenges (like structure-effectiveness-applicability relationship) for PEG/polysaccharides are proposed and addressed for food fields.
Collapse
Affiliation(s)
- Linshan Gong
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Juncheng Zhu
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Yuxin Yang
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Shihao Qiao
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Liang Ma
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Hongxia Wang
- College of Food Science, Southwest University, Chongqing 400715, PR China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China; Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, PR China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, PR China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China.
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing 400715, PR China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China; Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, PR China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, PR China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China.
| |
Collapse
|
15
|
Cai R, Shan Y, Du F, Miao Z, Zhu L, Hang L, Xiao L, Wang Z. Injectable hydrogels as promising in situ therapeutic platform for cartilage tissue engineering. Int J Biol Macromol 2024; 261:129537. [PMID: 38278383 DOI: 10.1016/j.ijbiomac.2024.129537] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Injectable hydrogels are gaining prominence as a biocompatible, minimally invasive, and adaptable platform for cartilage tissue engineering. Commencing with their synthesis, this review accentuates the tailored matrix formulations and cross-linking techniques essential for fostering three-dimensional cell culture and melding with complex tissue structures. Subsequently, it spotlights the hydrogels' enhanced properties, highlighting their augmented functionalities and broadened scope in cartilage tissue repair applications. Furthermore, future perspectives are advocated, urging continuous innovation and exploration to surmount existing challenges and harness the full clinical potential of hydrogels in regenerative medicine. Such advancements are crucial for validating the long-term efficacy and safety of hydrogels, positioning them as a promising direction in regenerative medicine to address cartilage-related ailments.
Collapse
Affiliation(s)
- Rong Cai
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Yisi Shan
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Fengyi Du
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212013, China
| | - Zhiwei Miao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Like Zhu
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Li Hang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| |
Collapse
|
16
|
Kong P, Liu X, Li Z, Wang J, Gao R, Feng S, Li H, Zhang F, Feng Z, Huang P, Wang S, Zhuang D, Ouyang W, Wang W, Pan X. Biodegradable Cardiac Occluder with Surface Modification by Gelatin-Peptide Conjugate to Promote Endogenous Tissue Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305967. [PMID: 37984880 PMCID: PMC10787076 DOI: 10.1002/advs.202305967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/20/2023] [Indexed: 11/22/2023]
Abstract
Transcatheter intervention has been the preferred treatment for congenital structural heart diseases by implanting occluders into the heart defect site through minimally invasive access. Biodegradable polymers provide a promising alternative for cardiovascular implants by conferring therapeutic function and eliminating long-term complications, but inducing in situ cardiac tissue regeneration remains a substantial clinical challenge. PGAG (polydioxanone/poly (l-lactic acid)-gelatin-A5G81) occluders are prepared by covalently conjugating biomolecules composed of gelatin and layer adhesive protein-derived peptides (A5G81) to the surface of polydioxanone and poly (l-lactic acid) fibers. The polymer microfiber-biomacromolecule-peptide frame with biophysical and biochemical cues could orchestrate the biomaterial-host cell interactions, by recruiting endogenous endothelial cells, promoting their adhesion and proliferation, and polarizing immune cells into anti-inflammatory phenotypes and augmenting the release of reparative cytokines. In a porcine atrial septal defect (ASD) model, PGAG occluders promote in situ tissue regeneration by accelerating surface endothelialization and regulating immune response, which mitigate inflammation and fibrosis formation, and facilitate the fusion of occluder with surrounding heart tissue. Collectively, this work highlights the modulation of cell-biomaterial interactions for tissue regeneration in cardiac defect models, ensuring endothelialization and extracellular matrix remodeling on polymeric scaffolds. Bioinspired cell-material interface offers a highly efficient and generalized approach for constructing bioactive coatings on medical devices.
Collapse
Affiliation(s)
- Pengxu Kong
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
| | - Xiang Liu
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Zefu Li
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
| | - Jingrong Wang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Rui Gao
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Shuyi Feng
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
| | - Hang Li
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
| | - Fengwen Zhang
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
| | - Zujian Feng
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Shouzheng Wang
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
- Key Laboratory of Innovative Cardiovascular DevicesChinese Academy of Medical SciencesBeijing100037China
| | - Donglin Zhuang
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
| | - Wenbin Ouyang
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
- Key Laboratory of Innovative Cardiovascular DevicesChinese Academy of Medical SciencesBeijing100037China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
- Key Laboratory of Innovative Cardiovascular DevicesChinese Academy of Medical SciencesBeijing100037China
| | - Xiangbin Pan
- Department of Structural Heart DiseaseNational Center for Cardiovascular DiseaseChina & State Key Laboratory of Cardiovascular DiseaseFuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNational Health Commission Key Laboratory of Cardiovascular Regeneration MedicineNational Clinical Research Center for Cardiovascular DiseasesBeijing100037China
- Key Laboratory of Innovative Cardiovascular DevicesChinese Academy of Medical SciencesBeijing100037China
| |
Collapse
|
17
|
Peyton SR. Engineering the endometrium. MED 2023; 4:495-496. [PMID: 37572649 DOI: 10.1016/j.medj.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
Endometrial tissue is a dynamic environment that regenerates alongside the menstrual cycle and in response to sex hormones. This month in Med, Gnecco and colleagues1 present an innovative, synthetic model of endometrial environment that supports long-term cultures, simulates a 28-day menstrual cycle, and could be employed to investigate reproductive pathophysiology.
Collapse
Affiliation(s)
- Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA 01003, USA; Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA; Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
18
|
Northcutt LA, Questell AM, Rhoades J, Rafat M. Development of an alginate-Matrigel hydrogel system to evaluate cancer cell behavior in the stiffness range of the bone marrow. FRONTIERS IN BIOMATERIALS SCIENCE 2023; 2:1140641. [PMID: 38169992 PMCID: PMC10760992 DOI: 10.3389/fbiom.2023.1140641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Bone metastasis is highly prevalent in breast cancer patients with metastatic disease. These metastatic cells may eventually form osteolytic lesions and affect the integrity of the bone, causing pathological fractures and impairing patient quality of life. Although some mechanisms have been determined in the metastatic cascade to the bone, little is known about how the mechanical cues of the bone marrow microenvironment influence tumor cell growth and invasion once they have homed to the secondary site. The mechanical properties within the bone marrow range from 0.5 kPa in the sinusoidal region to 40 kPa in the endosteal region. Here, we report an alginate-Matrigel hydrogel that can be modulated to the stiffness range of the bone marrow and used to evaluate tumor cell behavior. We fabricated alginate-Matrigel hydrogels with varying calcium sulfate (CaSO4) concentrations to tune stiffness, and we demonstrated that these hydrogels recapitulated the mechanical properties observed in the bone marrow microenvironment (0.7-16 kPa). We encapsulated multiple breast cancer cell lines into these hydrogels to assess growth and invasion. Tumor cells in stiffer hydrogels exhibited increased proliferation and enhanced elongation compared to lower stiffness hydrogels, which suggests that stiffer environments in the bone marrow promote cellular invasive capacity. This work establishes a system that replicates bone marrow mechanical properties to elucidate the physical factors that contribute to metastatic growth.
Collapse
Affiliation(s)
- Logan A. Northcutt
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
| | - Alyssa M. Questell
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Julie Rhoades
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Marjan Rafat
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
19
|
Oliver-Cervelló L, Martin-Gómez H, Gonzalez-Garcia C, Salmeron-Sanchez M, Ginebra MP, Mas-Moruno C. Protease-degradable hydrogels with multifunctional biomimetic peptides for bone tissue engineering. Front Bioeng Biotechnol 2023; 11:1192436. [PMID: 37324414 PMCID: PMC10267393 DOI: 10.3389/fbioe.2023.1192436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Mimicking bone extracellular matrix (ECM) is paramount to develop novel biomaterials for bone tissue engineering. In this regard, the combination of integrin-binding ligands together with osteogenic peptides represents a powerful approach to recapitulate the healing microenvironment of bone. In the present work, we designed polyethylene glycol (PEG)-based hydrogels functionalized with cell instructive multifunctional biomimetic peptides (either with cyclic RGD-DWIVA or cyclic RGD-cyclic DWIVA) and cross-linked with matrix metalloproteinases (MMPs)-degradable sequences to enable dynamic enzymatic biodegradation and cell spreading and differentiation. The analysis of the intrinsic properties of the hydrogel revealed relevant mechanical properties, porosity, swelling and degradability to engineer hydrogels for bone tissue engineering. Moreover, the engineered hydrogels were able to promote human mesenchymal stem cells (MSCs) spreading and significantly improve their osteogenic differentiation. Thus, these novel hydrogels could be a promising candidate for applications in bone tissue engineering, such as acellular systems to be implanted and regenerate bone or in stem cells therapy.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Cristina Gonzalez-Garcia
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| |
Collapse
|
20
|
Ligorio C, Mata A. Synthetic extracellular matrices with function-encoding peptides. NATURE REVIEWS BIOENGINEERING 2023; 1:1-19. [PMID: 37359773 PMCID: PMC10127181 DOI: 10.1038/s44222-023-00055-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 06/28/2023]
Abstract
The communication of cells with their surroundings is mostly encoded in the epitopes of structural and signalling proteins present in the extracellular matrix (ECM). These peptide epitopes can be incorporated in biomaterials to serve as function-encoding molecules to modulate cell-cell and cell-ECM interactions. In this Review, we discuss natural and synthetic peptide epitopes as molecular tools to bioengineer bioactive hydrogel materials. We present a library of functional peptide sequences that selectively communicate with cells and the ECM to coordinate biological processes, including epitopes that directly signal to cells, that bind ECM components that subsequently signal to cells, and that regulate ECM turnover. We highlight how these epitopes can be incorporated in different biomaterials as individual or multiple signals, working synergistically or additively. This molecular toolbox can be applied in the design of biomaterials aimed at regulating or controlling cellular and tissue function, repair and regeneration.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
- School of Pharmacy, University of Nottingham, Nottingham, UK
| |
Collapse
|
21
|
Pushparaj K, Balasubramanian B, Pappuswamy M, Anand Arumugam V, Durairaj K, Liu WC, Meyyazhagan A, Park S. Out of Box Thinking to Tangible Science: A Benchmark History of 3D Bio-Printing in Regenerative Medicine and Tissues Engineering. Life (Basel) 2023; 13:954. [PMID: 37109483 PMCID: PMC10145662 DOI: 10.3390/life13040954] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023] Open
Abstract
Advancements and developments in the 3D bioprinting have been promising and have met the needs of organ transplantation. Current improvements in tissue engineering constructs have enhanced their applications in regenerative medicines and other medical fields. The synergistic effects of 3D bioprinting have brought technologies such as tissue engineering, microfluidics, integrated tissue organ printing, in vivo bioprinted tissue implants, artificial intelligence and machine learning approaches together. These have greatly impacted interventions in medical fields, such as medical implants, multi-organ-on-chip models, prosthetics, drug testing tissue constructs and much more. This technological leap has offered promising personalized solutions for patients with chronic diseases, and neurodegenerative disorders, and who have been in severe accidents. This review discussed the various standing printing methods, such as inkjet, extrusion, laser-assisted, digital light processing, and stereolithographic 3D bioprinter models, adopted for tissue constructs. Additionally, the properties of natural, synthetic, cell-laden, dECM-based, short peptides, nanocomposite and bioactive bioinks are briefly discussed. Sequels of several tissue-laden constructs such as skin, bone and cartilage, liver, kidney, smooth muscles, cardiac and neural tissues are briefly analyzed. Challenges, future perspectives and the impact of microfluidics in resolving the limitations in the field, along with 3D bioprinting, are discussed. Certainly, a technology gap still exists in the scaling up, industrialization and commercialization of this technology for the benefit of stakeholders.
Collapse
Affiliation(s)
- Karthika Pushparaj
- Department of Zoology, School of Biosciences, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641 043, Tamil Nadu, India;
| | | | - Manikantan Pappuswamy
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru 560 076, Karnataka, India
| | - Vijaya Anand Arumugam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Kaliannan Durairaj
- Department of Infection Biology, School of Medicine, Wonkwang University, lksan 54538, Republic of Korea
| | - Wen-Chao Liu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Arun Meyyazhagan
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru 560 076, Karnataka, India
| | - Sungkwon Park
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul 05006, Republic of Korea;
| |
Collapse
|
22
|
de Janon A, Mantalaris A, Panoskaltsis N. Three-Dimensional Human Bone Marrow Organoids for the Study and Application of Normal and Abnormal Hematoimmunopoiesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:895-904. [PMID: 36947817 PMCID: PMC7614371 DOI: 10.4049/jimmunol.2200836] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/18/2023] [Indexed: 03/24/2023]
Abstract
Hematoimmunopoiesis takes place in the adult human bone marrow (BM), which is composed of heterogeneous niches with complex architecture that enables tight regulation of homeostatic and stress responses. There is a paucity of representative culture systems that recapitulate the heterogeneous three-dimensional (3D) human BM microenvironment and that can endogenously produce soluble factors and extracellular matrix that deliver culture fidelity for the study of both normal and abnormal hematopoiesis. Native BM lymphoid populations are also poorly represented in current in vitro and in vivo models, creating challenges for the study and treatment of BM immunopathology. BM organoid models leverage normal 3D organ structure to recreate functional niche microenvironments. Our focus herein is to review the current state of the art in the use of 3D BM organoids, focusing on their capacities to recreate critical quality attributes of the in vivo BM microenvironment for the study of human normal and abnormal hematopoiesis.
Collapse
Affiliation(s)
- Alejandro de Janon
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, The Georgia Institute of Technology, Atlanta, GA, USA
| | - Athanasios Mantalaris
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, The Georgia Institute of Technology, Atlanta, GA, USA
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Ireland
- National Institute for Bioprocessing Research and Training, Ireland
| | - Nicki Panoskaltsis
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, The Georgia Institute of Technology, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Ireland
- Department of Haematology, St. James’s Hospital Dublin, Ireland
| |
Collapse
|
23
|
Ganguly K, Dutta SD, Randhawa A, Patel DK, Patil TV, Lim KT. Transcriptomic Changes toward Osteogenic Differentiation of Mesenchymal Stem Cells on 3D-Printed GelMA/CNC Hydrogel under Pulsatile Pressure Environment. Adv Healthc Mater 2023; 12:e2202163. [PMID: 36637340 DOI: 10.1002/adhm.202202163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/20/2022] [Indexed: 01/14/2023]
Abstract
Biomimetic soft hydrogels used in bone tissue engineering frequently produce unsatisfactory outcomes. Here, it is investigated how human bone-marrow-derived mesenchymal stem cells (hBMSCs) differentiated into early osteoblasts on remarkably soft 3D hydrogel (70 ± 0.00049 Pa). Specifically, hBMSCs seeded onto cellulose nanocrystals incorporated methacrylate gelatin hydrogels are subjected to pulsatile pressure stimulation (PPS) of 5-20 kPa for 7 days. The PPS stimulates cellular processes such as mechanotransduction, cytoskeletal distribution, prohibition of oxidative stress, calcium homeostasis, osteogenic marker gene expression, and osteo-specific cytokine secretions in hBMSCs on soft substrates. The involvement of Piezo 1 is the main ion channel involved in mechanotransduction. Additionally, RNA-sequencing results reveal differential gene expression concerning osteogenic differentiation, bone mineralization, ion channel activity, and focal adhesion. These findings suggest a practical and highly scalable method for promoting stem cell commitment to osteogenesis on soft matrices for clinical reconstruction.
Collapse
Affiliation(s)
- Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Dinesh K Patel
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Biomechagen Co., Ltd., Chuncheon, 24341, Republic of Korea
| |
Collapse
|
24
|
Peyton SR, Platt MO, Cukierman E. Challenges and Opportunities Modeling the Dynamic Tumor Matrisome. BME FRONTIERS 2023; 4:0006. [PMID: 37849664 PMCID: PMC10521682 DOI: 10.34133/bmef.0006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/28/2022] [Indexed: 10/19/2023] Open
Abstract
We need novel strategies to target the complexity of cancer and, particularly, of metastatic disease. As an example of this complexity, certain tissues are particularly hospitable environments for metastases, whereas others do not contain fertile microenvironments to support cancer cell growth. Continuing evidence that the extracellular matrix (ECM) of tissues is one of a host of factors necessary to support cancer cell growth at both primary and secondary tissue sites is emerging. Research on cancer metastasis has largely been focused on the molecular adaptations of tumor cells in various cytokine and growth factor environments on 2-dimensional tissue culture polystyrene plates. Intravital imaging, conversely, has transformed our ability to watch, in real time, tumor cell invasion, intravasation, extravasation, and growth. Because the interstitial ECM that supports all cells in the tumor microenvironment changes over time scales outside the possible window of typical intravital imaging, bioengineers are continuously developing both simple and sophisticated in vitro controlled environments to study tumor (and other) cell interactions with this matrix. In this perspective, we focus on the cellular unit responsible for upholding the pathologic homeostasis of tumor-bearing organs, cancer-associated fibroblasts (CAFs), and their self-generated ECM. The latter, together with tumoral and other cell secreted factors, constitute the "tumor matrisome". We share the challenges and opportunities for modeling this dynamic CAF/ECM unit, the tools and techniques available, and how the tumor matrisome is remodeled (e.g., via ECM proteases). We posit that increasing information on tumor matrisome dynamics may lead the field to alternative strategies for personalized medicine outside genomics.
Collapse
Affiliation(s)
- Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Manu O. Platt
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| |
Collapse
|
25
|
Long S, Xie C, Lu X. Natural polymer‐based adhesive hydrogel for biomedical applications. BIOSURFACE AND BIOTRIBOLOGY 2022. [DOI: 10.1049/bsb2.12036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Siyu Long
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| | - Chaoming Xie
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| | - Xiong Lu
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| |
Collapse
|
26
|
Ho TC, Chang CC, Chan HP, Chung TW, Shu CW, Chuang KP, Duh TH, Yang MH, Tyan YC. Hydrogels: Properties and Applications in Biomedicine. Molecules 2022; 27:2902. [PMID: 35566251 PMCID: PMC9104731 DOI: 10.3390/molecules27092902] [Citation(s) in RCA: 280] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 12/19/2022] Open
Abstract
Hydrogels are crosslinked polymer chains with three-dimensional (3D) network structures, which can absorb relatively large amounts of fluid. Because of the high water content, soft structure, and porosity of hydrogels, they closely resemble living tissues. Research in recent years shows that hydrogels have been applied in various fields, such as agriculture, biomaterials, the food industry, drug delivery, tissue engineering, and regenerative medicine. Along with the underlying technology improvements of hydrogel development, hydrogels can be expected to be applied in more fields. Although not all hydrogels have good biodegradability and biocompatibility, such as synthetic hydrogels (polyvinyl alcohol, polyacrylamide, polyethylene glycol hydrogels, etc.), their biodegradability and biocompatibility can be adjusted by modification of their functional group or incorporation of natural polymers. Hence, scientists are still interested in the biomedical applications of hydrogels due to their creative adjustability for different uses. In this review, we first introduce the basic information of hydrogels, such as structure, classification, and synthesis. Then, we further describe the recent applications of hydrogels in 3D cell cultures, drug delivery, wound dressing, and tissue engineering.
Collapse
Affiliation(s)
- Tzu-Chuan Ho
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
| | - Chin-Chuan Chang
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Electrical Engineering, I-Shou University, Kaohsiung 840, Taiwan
| | - Hung-Pin Chan
- Department of Nuclear Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan;
| | - Tze-Wen Chung
- Biomedical Engineering Research and Development Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Chih-Wen Shu
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
| | - Kuo-Pin Chuang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
| | - Tsai-Hui Duh
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hui Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Center of General Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821, Taiwan
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
27
|
Gilchrist AE, Harley BA. Engineered Tissue Models to Replicate Dynamic Interactions within the Hematopoietic Stem Cell Niche. Adv Healthc Mater 2022; 11:e2102130. [PMID: 34936239 PMCID: PMC8986554 DOI: 10.1002/adhm.202102130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cells are the progenitors of the blood and immune system and represent the most widely used regenerative therapy. However, their rarity and limited donor base necessitate the design of ex vivo systems that support HSC expansion without the loss of long-term stem cell activity. This review describes recent advances in biomaterials systems to replicate features of the hematopoietic niche. Inspired by the native bone marrow, these instructive biomaterials provide stimuli and cues from cocultured niche-associated cells to support HSC encapsulation and expansion. Engineered systems increasingly enable study of the dynamic nature of the matrix and biomolecular environment as well as the role of cell-cell signaling (e.g., autocrine feedback vs paracrine signaling between dissimilar cells). The inherent coupling of material properties, biotransport of cell-secreted factors, and cell-mediated remodeling motivate dynamic biomaterial systems as well as characterization and modeling tools capable of evaluating a temporally evolving tissue microenvironment. Recent advances in HSC identification and tracking, model-based experimental design, and single-cell culture platforms facilitate the study of the effect of constellations of matrix, cell, and soluble factor signals on HSC fate. While inspired by the HSC niche, these tools are amenable to the broader stem cell engineering community.
Collapse
Affiliation(s)
- Aidan E. Gilchrist
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|