1
|
Mendes M, Morais AS, Carlos A, Sousa JJ, Pais AC, Mihăilă SM, Vitorino C. Organ-on-a-chip: Quo vademus? Applications and regulatory status. Colloids Surf B Biointerfaces 2025; 249:114507. [PMID: 39826309 DOI: 10.1016/j.colsurfb.2025.114507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/15/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Organ-on-a-chip systems, also referred to as microphysiological systems (MPS), represent an advance in bioengineering microsystems designed to mimic key aspects of human organ physiology and function. Drawing inspiration from the intricate and hierarchical architecture of the human body, these innovative platforms have emerged as invaluable in vitro tools with wide-ranging applications in drug discovery and development, as well as in enhancing our understanding of disease physiology. The facility to replicate human tissues within physiologically relevant three-dimensional multicellular environments empowers organ-on-a-chip systems with versatility throughout different stages of the drug development process. Moreover, these systems can be tailored to mimic specific disease states, facilitating the investigation of disease progression, drug responses, and potential therapeutic interventions. In particular, they can demonstrate, in early-phase pre-clinical studies, the safety and toxicity profiles of potential therapeutic compounds. Furthermore, they play a pivotal role in the in vitro evaluation of drug efficacy and the modeling of human diseases. One of the most promising prospects of organ-on-a-chip technology is to simulate the pathophysiology of specific subpopulations and even individual patients, thereby being used in personalized medicine. By mimicking the physiological responses of diverse patient groups, these systems hold the promise of revolutionizing therapeutic strategies, guiding them towards tailored intervention to the unique needs of each patient. This review presents the development status and evolution of microfluidic platforms that have facilitated the transition from cells to organs recreated on chips and some of the opportunities and applications offered by organ-on-a-chip technology. Additionally, the current potential and future perspectives of these microphysiological systems and the challenges this technology still faces are discussed.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal
| | - Ana Sofia Morais
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Carlos
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - João José Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal
| | - Alberto Canelas Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal
| | - Silvia M Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal.
| |
Collapse
|
2
|
Artamonov MY, Sokov EL, Kornilova LE, Minenko IA. The Intraosseous Environment: Physiological Parameters, Regulatory Mechanisms, and Emerging Insights in Bone Biology. Int J Mol Sci 2025; 26:3876. [PMID: 40332558 PMCID: PMC12027868 DOI: 10.3390/ijms26083876] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
The intraosseous environment is a dynamic and intricate system integral to bone health, encompassing vascular, cellular, and biochemical interactions that drive key processes such as hematopoiesis, bone remodeling, and systemic mineral regulation. This review examines the structural composition of the bone matrix, the diverse cellular landscape, and the interconnected vascular and nervous networks, highlighting their roles in preserving bone function and responding to pathological changes. Recent studies reveal regulatory mechanisms involving oxygen tension, ionic balance, signaling molecules, and mechanotransduction pathways that shape bone metabolism and its adaptation to mechanical forces. Insights into the bone microenvironment's metabolic shifts in cancer and its interaction with inflammation underscore its pivotal role in disease progression and therapeutic innovation. Additionally, advances in imaging techniques and biomaterials fuel progress in bone regeneration and understanding its microenvironment. Exploring the intricate physiochemical dynamics and regulatory networks within the intraosseous system unlocks potential clinical applications in bone diseases, tissue engineering, and systemic metabolic disorders. This comprehensive review bridges fundamental science with translational research, offering insights into the complex yet essential intraosseous environment.
Collapse
Affiliation(s)
- Mikhail Yu. Artamonov
- Department of Physical Medicine and Rehabilitation, Penn Medicine Princeton Health, Plainsboro, NJ 08536, USA
| | - Evgeniy L. Sokov
- Department of Algology, Peoples’ Friendship University, Moscow 117198, Russia
| | | | - Inessa A. Minenko
- Department of Sports Medicine, Sechenov Medical University, Moscow 119991, Russia;
| |
Collapse
|
3
|
Shi J, Wan Y, Jia H, Skeldon G, Jan Cornelissen D, Wesencraft K, Wu J, McConnell G, Chen Q, Liu D, Shu W. Printing Cell Embedded Sacrificial Strategy for Microvasculature using Degradable DNA Biolubricant. Angew Chem Int Ed Engl 2025; 64:e202417510. [PMID: 39460720 PMCID: PMC11914955 DOI: 10.1002/anie.202417510] [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: 10/04/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 10/28/2024]
Abstract
Microvasculature is essential for the continued function of cells in tissue and is fundamental in the fields of tissue engineering, organ repair and drug screening. However, the fabrication of microvasculature is still challenging using existing strategies. Here, we developed a general PRINting Cell Embedded Sacrificial Strategy (PRINCESS) and successfully fabricated microvasculatures using degradable DNA biolubricant. This is the first demonstration of direct cell printing to fabricate microvasculature, which eliminates the need for a subsequent cell seeding process and the associated deficiencies. Utilizing the shear-thinning property of DNA hydrogels as a novel sacrificial, cell-laden biolubricant, we can print a 70 μm endothelialized microvasculature, breaking the limit of 100 μm. To our best knowledge, this is the smallest endothelialized microvasculature that has ever been bioprinted so far. In addition, the self-healing property of DNA hydrogels allows the creation of continuous branched structures. This strategy provides a new platform for constructing complex hierarchical vascular networks and offers new opportunity towards engineering thick tissues. The extremely low volume of sacrificial biolubricant paves the way for DNA hydrogels to be used in practical tissue engineering applications. The high-resolution bioprinting technique also exhibits great potential for printing lymphatics, retinas and neural networks in the future.
Collapse
Affiliation(s)
- Jiezhong Shi
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Department of Chemistry Tsinghua UniversityBeijing100084China
- Department of Biomedical EngineeringUniversity of StrathclydeGlasgowG4 0NWUnited Kingdom
- SINOPEC Key Laboratory of Research and Application of Medical and Hygienic MaterialsSINOPEC Beijing Research Institute of Chemical Industry Co., Ltd.Beijing, 100013China
| | - Yifei Wan
- Department of Biomedical EngineeringUniversity of StrathclydeGlasgowG4 0NWUnited Kingdom
| | - Haoyang Jia
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Department of Chemistry Tsinghua UniversityBeijing100084China
| | - Gregor Skeldon
- Department of Biomedical EngineeringUniversity of StrathclydeGlasgowG4 0NWUnited Kingdom
| | - Dirk Jan Cornelissen
- Department of Biomedical EngineeringUniversity of StrathclydeGlasgowG4 0NWUnited Kingdom
| | - Katrina Wesencraft
- Department of Physics, SUPAUniversity of StrathclydeGlasgowG4 0NGUnited Kingdom
| | - Junxi Wu
- Department of Biomedical EngineeringUniversity of StrathclydeGlasgowG4 0NWUnited Kingdom
| | - Gail McConnell
- Department of Physics, SUPAUniversity of StrathclydeGlasgowG4 0NGUnited Kingdom
| | - Quan Chen
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied Chemistry, Chinese Academy of SciencesChangchun130022China
| | - Dongsheng Liu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Department of Chemistry Tsinghua UniversityBeijing100084China
| | - Wenmiao Shu
- Department of Biomedical EngineeringUniversity of StrathclydeGlasgowG4 0NWUnited Kingdom
| |
Collapse
|
4
|
Kozalak G, Koşar A. Bone-on-a-Chip Systems for Hematological Cancers. BIOSENSORS 2025; 15:176. [PMID: 40136973 PMCID: PMC11940066 DOI: 10.3390/bios15030176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Hematological malignancies originating from blood, bone marrow, and lymph nodes include leukemia, lymphoma, and myeloma, which necessitate the use of a distinct chemotherapeutic approach. Drug resistance frequently complicates their treatment, highlighting the need for predictive tools to guide therapeutic decisions. Conventional 2D/3D cell cultures do not fully encompass in vivo criteria, and translating disease models from mice to humans proves challenging. Organ-on-a-chip technology presents an avenue to surmount genetic disparities between species, offering precise design, concurrent manipulation of various cell types, and extrapolation of data to human physiology. The development of bone-on-a-chip (BoC) systems is crucial for accurately representing the in vivo bone microenvironment, predicting drug responses for hematological cancers, mitigating drug resistance, and facilitating personalized therapeutic interventions. BoC systems for modeling hematological cancers and drug research can encompass intricate designs and integrated platforms for analyzing drug response data to simulate disease scenarios. This review provides a comprehensive examination of BoC systems applicable to modeling hematological cancers and visualizing drug responses within the intricate context of bone. It thoroughly discusses the materials pertinent to BoC systems, suitable in vitro techniques, the predictive capabilities of BoC systems in clinical settings, and their potential for commercialization.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
5
|
Verdugo-Avello F, Wychowaniec JK, Villacis-Aguirre CA, D'Este M, Toledo JR. Bone microphysiological models for biomedical research. LAB ON A CHIP 2025; 25:806-836. [PMID: 39906932 DOI: 10.1039/d4lc00762j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Bone related disorders are highly prevalent, and many of these pathologies still do not have curative and definitive treatment methods. This is due to a complex interplay of multiple factors, such as the crosstalk between different tissues and cellular components, all of which are affected by microenvironmental factors. Moreover, these bone pathologies are specific, and current treatment results vary from patient to patient owing to their intrinsic biological variability. Current approaches in drug development to deliver new drug candidates against common bone disorders, such as standard two-dimensional (2D) cell culture and animal-based studies, are now being replaced by more relevant diseases modelling, such as three-dimension (3D) cell culture and primary cells under human-focused microphysiological systems (MPS) that can resemble human physiology by mimicking 3D tissue organization and cell microenvironmental cues. In this review, various technological advancements for in vitro bone modeling are discussed, highlighting the progress in biomaterials used as extracellular matrices, stem cell biology, and primary cell culture techniques. With emphasis on examples of modeling healthy and disease-associated bone tissues, this tutorial review aims to survey current approaches of up-to-date bone-on-chips through MPS technology, with special emphasis on the scaffold and chip capabilities for mimicking the bone extracellular matrix as this is the key environment generated for cell crosstalk and interaction. The relevant bone models are studied with critical analysis of the methods employed, aiming to serve as a tool for designing new and translational approaches. Additionally, the features reported in these state-of-the-art studies will be useful for modeling bone pathophysiology, guiding future improvements in personalized bone models that can accelerate drug discovery and clinical translation.
Collapse
Affiliation(s)
- Francisco Verdugo-Avello
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | | | - Carlos A Villacis-Aguirre
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Jorge R Toledo
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| |
Collapse
|
6
|
Ghobadi F, Saadatmand M, Simorgh S, Brouki Milan P. Microfluidic 3D cell culture: potential application of collagen hydrogels with an optimal dose of bioactive glasses. Sci Rep 2025; 15:569. [PMID: 39747624 PMCID: PMC11696724 DOI: 10.1038/s41598-024-84346-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
We engineered a microfluidic platform to study the effects of bioactive glass nanoparticles (BGNs) on cell viability under static culture. We incorporated different concentrations of BGNs (1%, 2%, and 3% w/v) in collagen hydrogel (with a concentration of 3.0 mg/mL). The microfluidic chip's dimensions were optimized through fluid flow and mass transfer simulations. Collagen type I extracted from rat tail tendons was used as the main material, and BGNs synthesized by the sol-gel method were used to enhance the mechanical properties of the hydrogel. The extracted collagen was characterized using FTIR and SDS-PAGE, and BGNs were analyzed using XRD, FTIR, DLS, and FE-SEM/EDX. The structure of the collagen-BGNs hydrogels was examined using SEM, and their mechanical properties were determined using rheological analysis. The cytotoxicity of BGNs was assessed using the MTT assay, and the viability of fibroblast (L929) cells encapsulated in the collagen-BGNs hydrogel inside the microfluidic device was assessed using a live/dead assay. Based on all these test results, the L929 cells showed high cell viability in vitro and promising microenvironment mimicry in a microfluidic device. Collagen3-BGNs3 (Collagen 3 mg/mL + BGNs 3% (w/v)) was chosen as the most suitable sample for further research on a microfluidic platform.
Collapse
Affiliation(s)
- Faezeh Ghobadi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Saadatmand
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Sara Simorgh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies inMedicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies inMedicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Wang Y, Yung P, Lu G, Liu Y, Ding C, Mao C, Li ZA, Tuan RS. Musculoskeletal Organs-on-Chips: An Emerging Platform for Studying the Nanotechnology-Biology Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2401334. [PMID: 38491868 PMCID: PMC11733728 DOI: 10.1002/adma.202401334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Nanotechnology-based approaches are promising for the treatment of musculoskeletal (MSK) disorders, which present significant clinical burdens and challenges, but their clinical translation requires a deep understanding of the complex interplay between nanotechnology and MSK biology. Organ-on-a-chip (OoC) systems have emerged as an innovative and versatile microphysiological platform to replicate the dynamics of tissue microenvironment for studying nanotechnology-biology interactions. This review first covers recent advances and applications of MSK OoCs and their ability to mimic the biophysical and biochemical stimuli encountered by MSK tissues. Next, by integrating nanotechnology into MSK OoCs, cellular responses and tissue behaviors may be investigated by precisely controlling and manipulating the nanoscale environment. Analysis of MSK disease mechanisms, particularly bone, joint, and muscle tissue degeneration, and drug screening and development of personalized medicine may be greatly facilitated using MSK OoCs. Finally, future challenges and directions are outlined for the field, including advanced sensing technologies, integration of immune-active components, and enhancement of biomimetic functionality. By highlighting the emerging applications of MSK OoCs, this review aims to advance the understanding of the intricate nanotechnology-MSK biology interface and its significance in MSK disease management, and the development of innovative and personalized therapeutic and interventional strategies.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Patrick Yung
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Gang Lu
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Yuwei Liu
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- The First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhenGuangdong518037P. R. China
| | - Changhai Ding
- Clinical Research CentreZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510260China
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Zhong Alan Li
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Key Laboratory of Regenerative MedicineMinistry of EducationSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077P. R. China
- Shenzhen Research InstituteThe Chinese University of Hong KongShenzhen518172P. R. China
| | - Rocky S. Tuan
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| |
Collapse
|
8
|
Zhong C, Tang Z, Yu X, Wang L, Ren C, Qin L, Zhou P. Advances in the Construction and Application of Bone-on-a-Chip Based on Microfluidic Technologies. J Biomed Mater Res B Appl Biomater 2024; 112:e35502. [PMID: 39555794 DOI: 10.1002/jbm.b.35502] [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: 06/19/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Bone-on-a-chip (BOC) models that based on microfluidic technology have widely applied to understand bone physiology and the pathogenesis of related diseases. In this review, we provide an overview of bone biology and related diseases, explain the advantages and applications of microfluidic technology in the construction of BOC models, and summarize their progress in physiology, pathology, and drug development. Finally, we discussed the problems to be solved and the future directions of microfluidic technology and BOC platforms, so as to provide a reference for researchers to design better BOC models.
Collapse
Affiliation(s)
- Chang Zhong
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Zihui Tang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Xin Yu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Lu Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Chenyuan Ren
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Gansu Health Vocational College, Lanzhou, China
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Tronolone JJ, Mohamed N, Chaftari CP, Sun Y, Mathur T, Jain A. Engineering and Evaluating Vascularized Organotypic Spheroids On-Chip. Curr Protoc 2024; 4:e70058. [PMID: 39570194 DOI: 10.1002/cpz1.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Organotypic spheroids are evolving as a mainstream in vitro modeling platform, but it is crucial to integrate vascular tissue and perfusion for maintaining their longevity, stability, and physiological relevance. Current vascularization methods remain underdeveloped, and several protocols are poorly reproducible and are limited to use by a few select groups who have designed these methods. To achieve standardization, we offer a step-by-step guide to vascularize organotypic spheroids in case studies of pancreatic islets and cancer spheroids. Our systematic approach spans microfluidic chip design, spheroid fabrication, and vascularization techniques (vasculogenesis and angiogenesis) while describing critical tissue engineering methods. We also include additional insights and operating guidelines within our protocols that characterize and quantitate these models with molecular assays as well as our integrated computational algorithms of mass transport through formed capillary vessels. These protocols contribute to establishing reproducibility, standardization, and enhanced adoption by other contemporary organ-chip researchers, who want to engineer vascularized organoid-based microphysiological platforms. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Design and fabrication of microfluidic chips for vascularized spheroids Basic Protocol 2: Organotypic spheroid fabrication Basic Protocol 3: Vascularized spheroids on-chip Basic Protocol 4: Functionality assays Support Protocol 1: Cell Culture Support Protocol 2: Immunocytochemistry.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
| | - Nadin Mohamed
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
| | - Christopher P Chaftari
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, Bryan, Texas
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, Texas
| |
Collapse
|
10
|
Firatligil-Yildirir B, Bati-Ayaz G, Nonappa, Pesen-Okvur D, Yalcin-Ozuysal O. Invasion/chemotaxis- and extravasation-chip models for breast cancer bone metastasis. PLoS One 2024; 19:e0309285. [PMID: 39418263 PMCID: PMC11486417 DOI: 10.1371/journal.pone.0309285] [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: 06/05/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024] Open
Abstract
Bone is one of the most frequently targeted organs in metastatic cancers including the breast. Breast cancer bone metastasis often results in devastating outcomes as limited treatment options are currently available. Therefore, innovative methods are needed to provide earlier detection and thus better treatment and prognosis. Here, we present a new approach to model bone-like microenvironments to detect invasion and extravasation of breast cancer cells using invasion/chemotaxis (IC-) and extravasation (EX-) chips, respectively. Our results show that the behaviors of MDA-MB-231 breast cancer cells on IC- and EX-chip models correlate with their in vivo metastatic potential. Our culture model constitutes cell lines representing osteoblasts, bone marrow stromal cells, and monocytes embedded in three-dimensional (3D) collagen I-based extracellular matrices of varying composition and stiffness. We show that collagen I offers a better bone-like environment for bone cells and matrix composition and stiffness regulate the invasion of breast cancer cells. Using in situ contactless rheological measurements under cell culture conditions, we show that the presence of cells increased the stiffness values of the matrices up to 1200 Pa when monitored for five days. This suggests that the cellular composition has a significant effect on regulating matrix mechanical properties, which in turn contribute to the invasiveness. The platforms we present here enable the investigation of the underlying molecular mechanisms in breast cancer bone metastasis and provide the groundwork of developing preclinical tools for the prediction of bone metastasis risk.
Collapse
Affiliation(s)
- Burcu Firatligil-Yildirir
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkiye
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Gizem Bati-Ayaz
- Izmir Institute of Technology, Biotechnology and Bioengineering Graduate Program, Izmir, Turkiye
| | - Nonappa
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Devrim Pesen-Okvur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkiye
| | - Ozden Yalcin-Ozuysal
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkiye
| |
Collapse
|
11
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [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: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
12
|
Fois MG, van Griensven M, Giselbrecht S, Habibović P, Truckenmüller RK, Tahmasebi Birgani ZN. Mini-bones: miniaturized bone in vitro models. Trends Biotechnol 2024; 42:910-928. [PMID: 38493050 DOI: 10.1016/j.tibtech.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 03/18/2024]
Abstract
In bone tissue engineering (TE) and regeneration, miniaturized, (sub)millimeter-sized bone models have become a popular trend since they bring about physiological biomimicry, precise orchestration of concurrent stimuli, and compatibility with high-throughput setups and high-content imaging. They also allow efficient use of cells, reagents, materials, and energy. In this review, we describe the state of the art of miniaturized in vitro bone models, or 'mini-bones', describing these models based on their characteristics of (multi)cellularity and engineered extracellular matrix (ECM), and elaborating on miniaturization approaches and fabrication techniques. We analyze the performance of 'mini-bone' models according to their applications for studying basic bone biology or as regeneration models, disease models, and screening platforms, and provide an outlook on future trends, challenges, and opportunities.
Collapse
Affiliation(s)
- Maria Gabriella Fois
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Martijn van Griensven
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Roman K Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| | - Zeinab Niloofar Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| |
Collapse
|
13
|
Morais AS, Mendes M, Cordeiro MA, Sousa JJ, Pais AC, Mihăilă SM, Vitorino C. Organ-on-a-Chip: Ubi sumus? Fundamentals and Design Aspects. Pharmaceutics 2024; 16:615. [PMID: 38794277 PMCID: PMC11124787 DOI: 10.3390/pharmaceutics16050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
This review outlines the evolutionary journey from traditional two-dimensional (2D) cell culture to the revolutionary field of organ-on-a-chip technology. Organ-on-a-chip technology integrates microfluidic systems to mimic the complex physiological environments of human organs, surpassing the limitations of conventional 2D cultures. This evolution has opened new possibilities for understanding cell-cell interactions, cellular responses, drug screening, and disease modeling. However, the design and manufacture of microchips significantly influence their functionality, reliability, and applicability to different biomedical applications. Therefore, it is important to carefully consider design parameters, including the number of channels (single, double, or multi-channels), the channel shape, and the biological context. Simultaneously, the selection of appropriate materials compatible with the cells and fabrication methods optimize the chips' capabilities for specific applications, mitigating some disadvantages associated with these systems. Furthermore, the success of organ-on-a-chip platforms greatly depends on the careful selection and utilization of cell resources. Advances in stem cell technology and tissue engineering have contributed to the availability of diverse cell sources, facilitating the development of more accurate and reliable organ-on-a-chip models. In conclusion, a holistic perspective of in vitro cellular modeling is provided, highlighting the integration of microfluidic technology and meticulous chip design, which play a pivotal role in replicating organ-specific microenvironments. At the same time, the sensible use of cell resources ensures the fidelity and applicability of these innovative platforms in several biomedical applications.
Collapse
Affiliation(s)
- Ana Sofia Morais
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - Marta Agostinho Cordeiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - João J. Sousa
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - Alberto Canelas Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - Silvia M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, The Netherlands;
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| |
Collapse
|
14
|
Jang HJ, Yoon JK. The Role of Vasculature and Angiogenic Strategies in Bone Regeneration. Biomimetics (Basel) 2024; 9:75. [PMID: 38392121 PMCID: PMC10887147 DOI: 10.3390/biomimetics9020075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Bone regeneration is a complex process that involves various growth factors, cell types, and extracellular matrix components. A crucial aspect of this process is the formation of a vascular network, which provides essential nutrients and oxygen and promotes osteogenesis by interacting with bone tissue. This review provides a comprehensive discussion of the critical role of vasculature in bone regeneration and the applications of angiogenic strategies, from conventional to cutting-edge methodologies. Recent research has shifted towards innovative bone tissue engineering strategies that integrate vascularized bone complexes, recognizing the significant role of vasculature in bone regeneration. The article begins by examining the role of angiogenesis in bone regeneration. It then introduces various in vitro and in vivo applications that have achieved accelerated bone regeneration through angiogenesis to highlight recent advances in bone tissue engineering. This review also identifies remaining challenges and outlines future directions for research in vascularized bone regeneration.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
| |
Collapse
|
15
|
Ji X, Bei HP, Zhong G, Shao H, He X, Qian X, Zhang Y, Zhao X. Premetastatic Niche Mimicking Bone-On-A-Chip: A Microfluidic Platform to Study Bone Metastasis in Cancer Patients. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207606. [PMID: 37605335 DOI: 10.1002/smll.202207606] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 07/08/2023] [Indexed: 08/23/2023]
Abstract
Primary cancer modulates the bone microenvironment to sow the seeds of dormancy and metastasis in tumor cells, leading to multiple organ metastasis and death. In this study, 3D printing and bone-on-a-chip (BOC) are combined to develop a BOC platform that mimics the pre-metastatic niches (PMNs) and facilitates elucidation of the interactions between bone-resident cells and metastatic tumor cells under the influence of primary cancer. Photocrosslinkable gelatin methacrylate (GelMA) is used as a 3D culturing hydrogel to encapsulate cells, and circulate tumor culture medium (CM) adjacent to the hydrogel to verify the critical role of mesenchymal stem cells (MSCs) and osteoclasts (RAW264.7s). Three niches: the dormancy niche, the perivascular niche, and the "vicious cycle" niche, are devised to recapitulate bone metastasis in one chip with high cell viability and excellent nutrient exchange. With respect to tumor dormancy and reactivation, the invadopodia formation of A549 lung cancer cells in communication with MSCs and RAW264.7 via the cortactin pathway is researched. As a proof of concept, the functionality and practicality of the platform are demonstrated by analyzing the invadopodia formation and the influence of various cells, and the establishment of the dynamic niches paves the way to understanding PMN formation and related drug discovery.
Collapse
Affiliation(s)
- Xiongfa Ji
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Guoqing Zhong
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Hongwei Shao
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Xuecheng He
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Xin Qian
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Yu Zhang
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| |
Collapse
|
16
|
Ye S, Cao Q, Ni P, Xiong S, Zhong M, Yuan T, Shan J, Liang J, Fan Y, Zhang X. Construction of Microfluidic Chip Structure for Cell Migration Studies in Bioactive Ceramics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302152. [PMID: 37282789 DOI: 10.1002/smll.202302152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/16/2023] [Indexed: 06/08/2023]
Abstract
Cell migration is an essential bioactive ceramics property and critical for bone induction, clinical application, and mechanism research. Standardized cell migration detection methods have many limitations, including a lack of dynamic fluid circulation and the inability to simulate cell behavior in vivo. Microfluidic chip technology, which mimics the human microenvironment and provides controlled dynamic fluid cycling, has the potential to solve these questions and generate reliable models of cell migration in vitro. In this study, a microfluidic chip is reconstructed to integrate the bioactive ceramic into the microfluidic chip structure to constitute a ceramic microbridge microfluidic chip system. Migration differences in the chip system are measured. By combining conventional detection methods with new biotechnology to analyze the causes of cell migration differences, it is found that the concentration gradients of ions and proteins adsorbed on the microbridge materials are directly related to the occurrence of cell migration behavior, which is consistent with previous reports and demonstrates the effectiveness of the microfluidic chip model. This model provides in vivo environment simulation and controllability of input and output conditions superior to standardized cell migration detection methods. The microfluidic chip system provides a new approach to studying and evaluating bioactive ceramics.
Collapse
Affiliation(s)
- Sheng Ye
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Quanle Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Panxianzhi Ni
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Shuting Xiong
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Meng Zhong
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Tun Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
- Sichuan Testing Centre for Biomaterials and Medical Devices, Chengdu, Sichuan, 610064, China
| | - Jing Shan
- Department of Gastroenterology, the 3rd People's Hospital of Chengdu, Southwest Jiaotong University, Chengdu, Sichuan, 610064, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
- Sichuan Testing Centre for Biomaterials and Medical Devices, Chengdu, Sichuan, 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| |
Collapse
|
17
|
Fan X, Yan Y, Zhao L, Xu X, Dong Y, Sun W. Establishment of the multi-component bone-on-a-chip: to explore therapeutic potential of DNA aptamers on endothelial cells. Front Cell Dev Biol 2023; 11:1183163. [PMID: 37377731 PMCID: PMC10291622 DOI: 10.3389/fcell.2023.1183163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Background: Despite great efforts to develop microvascular bone chips in previous studies, current bone chips still lacked multi-component of human-derived cells close to human bone tissue. Bone microvascular endothelial cells (BMECs) were demonstrated to be closely related to the glucocorticoid (GC)-induced osteonecrosis of the femoral head (ONFH). Tumor necrosis factor-alpha (TNF-α) aptamer has been proved to bind to its receptor and block cascade activities. Objective: There are two main objectives in this study: 1) to establish a multi-component bone-on-a-chip within the microfluidic system in vitro, 2) to explore the therapeutic potential of TNF-α aptamer on BMECs in the GC-induced ONFH model. Methods: Histological features of clinical samples were analyzed before BMECs isolation. The functional bone-on-a-chip consists of the vascular channel, stromal channel and structure channel. GC-induced ONFH model was established based on the multi-component of human-derived cells. Truncation and dimerization were performed on a previously reported DNA aptamer (VR11). BMECs apoptosis, cytoskeleton and angiogenesis status in the ONFH model were observed by the TUNEL staining and confocal microscope. Results: The multi-component of BMECs, human embryonic lung fibroblasts and hydroxyapatite were cultured within the microfluidic bone-on-a-chip. TNF-α was found up-regulated in the necrotic regions of femoral heads in clinical samples and similar results were re-confirmed in the ONFH model established in the microfluidic platform by detecting cell metabolites. Molecular docking simulations indicated that the truncated TNF-α aptamer could improve the aptamer-protein interactions. Further results from the TUNEL staining and confocal microscopy showed that the truncated aptamer could protect BMECs from apoptosis and alleviate GC-induced damages to cytoskeleton and vascularization. Conclusion: In summary, a microfluidic multi-component bone-on-a-chip was established with 'off-chip' analysis of cell metabolism. GC-induced ONFH model was achieved based on the platform. Our findings provided initial evidence on the possible potentials of TNF-α aptamer as a new type of TNF-α inhibitor for patients with ONFH.
Collapse
Affiliation(s)
- Xiaoyu Fan
- Peking University Health Science Center, China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yuhan Yan
- Department of Pharmacy, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lianhui Zhao
- Department of Pharmacy, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xin Xu
- Peking Union Medical College, China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yiyang Dong
- Department of Pharmacy, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Wei Sun
- Peking University Health Science Center, China-Japan Friendship School of Clinical Medicine, Beijing, China
- Orthopedics Department, China-Japan Friendship Hospital, Beijing, China
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
18
|
Kim MK, Paek K, Woo SM, Kim JA. Bone-on-a-Chip: Biomimetic Models Based on Microfluidic Technologies for Biomedical Applications. ACS Biomater Sci Eng 2023. [PMID: 37183366 DOI: 10.1021/acsbiomaterials.3c00066] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
With the increasing importance of preclinical evaluation of newly developed drugs or treatments, in vitro organ or disease models are necessary. Although various organ-specific on-chip (organ-on-a-chip, or OOC) systems have been developed as emerging in vitro models, bone-on-a-chip (BOC) systems that recapitulate the bone microenvironment have been less developed or reviewed compared with other OOCs. The bone is one of the most dynamic organs and undergoes continuous remodeling throughout its lifetime. The aging population is growing worldwide, and healthcare costs are rising rapidly. Since in vitro BOC models that recapitulate native bone niches and pathological features can be important for studying the underlying mechanism of orthopedic diseases and predicting drug responses in preclinical trials instead of in animals, the development of biomimetic BOCs with high efficiency and fidelity will be accelerated further. Here, we review recently engineered BOCs developed using various microfluidic technologies and investigate their use to model the bone microenvironment. We have also explored various biomimetic strategies based on biological, geometrical, and biomechanical cues for biomedical applications of BOCs. Finally, we addressed the limitations and challenging issues of current BOCs that should be overcome to obtain more acceptable BOCs in the biomedical and pharmaceutical industries.
Collapse
Affiliation(s)
- Min Kyeong Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Kyurim Paek
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Program in Biomicro System Technology, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Mi Woo
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Republic of Korea
| |
Collapse
|
19
|
Li ZA, Sant S, Cho SK, Goodman SB, Bunnell BA, Tuan RS, Gold MS, Lin H. Synovial joint-on-a-chip for modeling arthritis: progress, pitfalls, and potential. Trends Biotechnol 2023; 41:511-527. [PMID: 35995600 PMCID: PMC9938846 DOI: 10.1016/j.tibtech.2022.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 12/30/2022]
Abstract
Disorders of the synovial joint, such as osteoarthritis (OA) and rheumatoid arthritis (RA), afflict a substantial proportion of the global population. However, current clinical management has not been focused on fully restoring the native function of joints. Organ-on-chip (OoC), also called a microphysiological system, which typically accommodates multiple human cell-derived tissues/organs under physiological culture conditions, is an emerging platform that potentially overcomes the limitations of current models in developing therapeutics. Herein, we review major steps in the generation of OoCs for studying arthritis, discuss the challenges faced when these novel platforms enter the next phase of development and application, and present the potential for OoC technology to investigate the pathogenesis of joint diseases and the development of efficacious therapies.
Collapse
Affiliation(s)
- Zhong Alan Li
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Sung Kwon Cho
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Stuart B Goodman
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rocky S Tuan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, China
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
20
|
Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G, Maruccio G. Organs-on-chips technologies – A guide from disease models to opportunities for drug development. Biosens Bioelectron 2023; 231:115271. [PMID: 37060819 DOI: 10.1016/j.bios.2023.115271] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 11/24/2022] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Current in-vitro 2D cultures and animal models present severe limitations in recapitulating human physiopathology with striking discrepancies in estimating drug efficacy and side effects when compared to human trials. For these reasons, microphysiological systems, organ-on-chip and multiorgans microdevices attracted considerable attention as novel tools for high-throughput and high-content research to achieve an improved understanding of diseases and to accelerate the drug development process towards more precise and eventually personalized standards. This review takes the form of a guide on this fast-growing field, providing useful introduction to major themes and indications for further readings. We start analyzing Organs-on-chips (OOC) technologies for testing the major drug administration routes: (1) oral/rectal route by intestine-on-a-chip, (2) inhalation by lung-on-a-chip, (3) transdermal by skin-on-a-chip and (4) intravenous through vascularization models, considering how drugs penetrate in the bloodstream and are conveyed to their targets. Then, we focus on OOC models for (other) specific organs and diseases: (1) neurodegenerative diseases with brain models and blood brain barriers, (2) tumor models including their vascularization, organoids/spheroids, engineering and screening of antitumor drugs, (3) liver/kidney on chips and multiorgan models for gastrointestinal diseases and metabolic assessment of drugs and (4) biomechanical systems recapitulating heart, muscles and bones structures and related diseases. Successively, we discuss technologies and materials for organ on chips, analyzing (1) microfluidic tools for organs-on-chips, (2) sensor integration for real-time monitoring, (3) materials and (4) cell lines for organs on chips. (Nano)delivery approaches for therapeutics and their on chip assessment are also described. Finally, we conclude with a critical discussion on current significance/relevance, trends, limitations, challenges and future prospects in terms of revolutionary impact on biomedical research, preclinical models and drug development.
Collapse
Affiliation(s)
- Anna Grazia Monteduro
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Silvia Rizzato
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Giusi Caragnano
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Giuseppe Maruccio
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy.
| |
Collapse
|
21
|
Zhang Y, Yu T, Ding J, Li Z. Bone-on-a-chip platforms and integrated biosensors: Towards advanced in vitro bone models with real-time biosensing. Biosens Bioelectron 2023; 219:114798. [PMID: 36257118 DOI: 10.1016/j.bios.2022.114798] [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: 07/03/2022] [Revised: 08/25/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Bone diseases, such as osteoporosis and bone defects, often lead to structural and functional deformities of the patient's body. Understanding the complicated pathophysiology and finding new drugs for bone diseases are in dire need but challenging with the conventional cell and animal models. Bone-on-a-chip (BoC) models recapitulate key features of bone at an unprecedented level and can potentially shift the paradigm of future bone research and therapeutic development. Nevertheless, current BoC models predominantly rely on off-chip analysis which provides only endpoint measurements. To this end, integrating biosensors within the BoC can provide non-invasive, continuous monitoring of the experiment progression, significantly facilitating bone research. This review aims to summarize research progress in BoC and biosensor integrations and share perspectives on this exciting but rudimentary research area. We first introduce the research progress of BoC models in the study of bone remodeling and bone diseases, respectively. We then summarize the need for BoC characterization and reported works on biosensor integration in organ chips. Finally, we discuss the limitations and future directions of BoC models and biosensor integrations as next-generation technologies for bone research.
Collapse
Affiliation(s)
- Yang Zhang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, 518060, China; School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Taozhao Yu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jingyi Ding
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Zida Li
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
22
|
Hrynevich A, Li Y, Cedillo-Servin G, Malda J, Castilho M. (Bio)fabrication of microfluidic devices and organs-on-a-chip. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
23
|
Petta D, D'Amora U, D'Arrigo D, Tomasini M, Candrian C, Ambrosio L, Moretti M. Musculoskeletal tissues-on-a-chip: role of natural polymers in reproducing tissue-specific microenvironments. Biofabrication 2022; 14. [PMID: 35931043 DOI: 10.1088/1758-5090/ac8767] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/05/2022] [Indexed: 11/12/2022]
Abstract
Over the past years, 3D in vitro models have been widely employed in the regenerative medicine field. Among them, organ-on-a-chip technology has the potential to elucidate cellular mechanism exploiting multichannel microfluidic devices to establish 3D co-culture systems that offer control over the cellular, physico-chemical and biochemical microenvironments. To deliver the most relevant cues to cells, it is of paramount importance to select the most appropriate matrix for mimicking the extracellular matrix of the native tissue. Natural polymers-based hydrogels are the elected candidates for reproducing tissue-specific microenvironments in musculoskeletal tissue-on-a-chip models owning to their interesting and peculiar physico-chemical, mechanical and biological properties. Despite these advantages, there is still a gap between the biomaterials complexity in conventional tissue engineering and the application of these biomaterials in 3D in vitro microfluidic models. In this review, the aim is to suggest the adoption of more suitable biomaterials, alternative crosslinking strategies and tissue engineered-inspired approaches in organ-on-a-chip to better mimic the complexity of physiological musculoskeletal tissues. Accordingly, after giving an overview of the musculoskeletal tissue compositions, the properties of the main natural polymers employed in microfluidic systems are investigated, together with the main musculoskeletal tissues-on-a-chip devices.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologis Lab, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Ugo D'Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, V.le J.F. Kennedy 54 Mostra d'Oltremare Pad 20, Naples, 80125, ITALY
| | - Daniele D'Arrigo
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Marta Tomasini
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco chies 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Christian Candrian
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, via Tesserete 46, Lugano, 6900, SWITZERLAND
| | - Luigi Ambrosio
- Institute of Polymers Composites and Biomaterials National Research Council, Viale Kennedy, Pozzuoli, Campania, 80078, ITALY
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| |
Collapse
|
24
|
Zhou J, Nie Y, Jin C, Zhang JXJ. Engineering Biomimetic Extracellular Matrix with Silica Nanofibers: From 1D Material to 3D Network. ACS Biomater Sci Eng 2022; 8:2258-2280. [PMID: 35377596 DOI: 10.1021/acsbiomaterials.1c01525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomaterials at nanoscale is a fast-expanding research field with which extensive studies have been conducted on understanding the interactions between cells and their surrounding microenvironments as well as intracellular communications. Among many kinds of nanoscale biomaterials, mesoporous fibrous structures are especially attractive as a promising approach to mimic the natural extracellular matrix (ECM) for cell and tissue research. Silica is a well-studied biocompatible, natural inorganic material that can be synthesized as morpho-genetically active scaffolds by various methods. This review compares silica nanofibers (SNFs) to other ECM materials such as hydrogel, polymers, and decellularized natural ECM, summarizes fabrication techniques for SNFs, and discusses different strategies of constructing ECM using SNFs. In addition, the latest progress on SNFs synthesis and biomimetic ECM substrates fabrication is summarized and highlighted. Lastly, we look at the wide use of SNF-based ECM scaffolds in biological applications, including stem cell regulation, tissue engineering, drug release, and environmental applications.
Collapse
Affiliation(s)
- Junhu Zhou
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Yuan Nie
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Congran Jin
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - John X J Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| |
Collapse
|
25
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
26
|
Neto E, Monteiro AC, Leite Pereira C, Simões M, Conde JP, Chu V, Sarmento B, Lamghari M. Micropathological Chip Modeling the Neurovascular Unit Response to Inflammatory Bone Condition. Adv Healthc Mater 2022; 11:e2102305. [PMID: 35158409 PMCID: PMC11468530 DOI: 10.1002/adhm.202102305] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Organ-on-a-chip in vitro platforms accurately mimic complex microenvironments offering the ability to recapitulate and dissect mechanisms of physiological and pathological settings, revealing their major importance to develop new therapeutic targets. Bone diseases, such as osteoarthritis, are extremely complex, comprising of the action of inflammatory mediators leading to unbalanced bone homeostasis and de-regulation of sensory innervation and angiogenesis. Although there are models to mimic bone vascularization or innervation, in vitro platforms merging the complexity of bone, vasculature, innervation, and inflammation are missing. Therefore, in this study a microfluidic-based neuro-vascularized bone chip (NVB chip) is proposed to 1) model the mechanistic interactions between innervation and angiogenesis in the inflammatory bone niche, and 2) explore, as a screening tool, novel strategies targeting inflammatory diseases, using a nano-based drug delivery system. It is possible to set the design of the platform and achieve the optimized conditions to address the neurovascular network under inflammation. Moreover, this system is validated by delivering anti-inflammatory drug-loaded nanoparticles to counteract the neuronal growth associated with pain perception. This reliable in vitro tool will allow understanding the bone neurovascular system, enlightening novel mechanisms behind the inflammatory bone diseases, bone destruction, and pain opening new avenues for new therapies discovery.
Collapse
Affiliation(s)
- Estrela Neto
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Ana Carolina Monteiro
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Catarina Leite Pereira
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Miguel Simões
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - João Pedro Conde
- Instituto de Engenharia de Sistemas e Computadores (INESC)Microsystems and NanotechnologiesRua Alves Redol, 91000‐029LisboaPortugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas e Computadores (INESC)Microsystems and NanotechnologiesRua Alves Redol, 91000‐029LisboaPortugal
| | - Bruno Sarmento
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- CESPUInstituto de Investigação e Formação Avançada em Ciências e Tecnologias da SaúdeRua Central da Gandra, 137Gandra4585‐116Portugal
| | - Meriem Lamghari
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| |
Collapse
|
27
|
Mykuliak A, Yrjänäinen A, Mäki AJ, Gebraad A, Lampela E, Kääriäinen M, Pakarinen TK, Kallio P, Miettinen S, Vuorenpää H. Vasculogenic Potency of Bone Marrow- and Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells Results in Differing Vascular Network Phenotypes in a Microfluidic Chip. Front Bioeng Biotechnol 2022; 10:764237. [PMID: 35211462 PMCID: PMC8861308 DOI: 10.3389/fbioe.2022.764237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/11/2022] [Indexed: 12/27/2022] Open
Abstract
The vasculature is an essential, physiological element in virtually all human tissues. Formation of perfusable vasculature is therefore crucial for reliable tissue modeling. Three-dimensional vascular networks can be formed through the co-culture of endothelial cells (ECs) with stromal cells embedded in hydrogel. Mesenchymal stem/stromal cells (MSCs) derived from bone marrow (BMSCs) and adipose tissue (ASCs) are an attractive choice as stromal cells due to their natural perivascular localization and ability to support formation of mature and stable microvessels in vitro. So far, BMSCs and ASCs have been compared as vasculature-supporting cells in static cultures. In this study, BMSCs and ASCs were co-cultured with endothelial cells in a fibrin hydrogel in a perfusable microfluidic chip. We demonstrated that using MSCs of different origin resulted in vascular networks with distinct phenotypes. Both types of MSCs supported formation of mature and interconnected microvascular networks-on-a-chip. However, BMSCs induced formation of fully perfusable microvasculature with larger vessel area and length whereas ASCs resulted in partially perfusable microvascular networks. Immunostainings revealed that BMSCs outperformed ASCs in pericytic characteristics. Moreover, co-culture with BMSCs resulted in significantly higher expression levels of endothelial and pericyte-specific genes, as well as genes involved in vasculature maturation. Overall, our study provides valuable knowledge on the properties of MSCs as vasculature-supporting cells and highlights the importance of choosing the application-specific stromal cell source for vascularized organotypic models.
Collapse
Affiliation(s)
- Anastasiia Mykuliak
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Antti-Juhana Mäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Arjen Gebraad
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Ella Lampela
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Minna Kääriäinen
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
28
|
Lipreri MV, Baldini N, Graziani G, Avnet S. Perfused Platforms to Mimic Bone Microenvironment at the Macro/Milli/Microscale: Pros and Cons. Front Cell Dev Biol 2022; 9:760667. [PMID: 35047495 PMCID: PMC8762164 DOI: 10.3389/fcell.2021.760667] [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: 08/18/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
As life expectancy increases, the population experiences progressive ageing. Ageing, in turn, is connected to an increase in bone-related diseases (i.e., osteoporosis and increased risk of fractures). Hence, the search for new approaches to study the occurrence of bone-related diseases and to develop new drugs for their prevention and treatment becomes more pressing. However, to date, a reliable in vitro model that can fully recapitulate the characteristics of bone tissue, either in physiological or altered conditions, is not available. Indeed, current methods for modelling normal and pathological bone are poor predictors of treatment outcomes in humans, as they fail to mimic the in vivo cellular microenvironment and tissue complexity. Bone, in fact, is a dynamic network including differently specialized cells and the extracellular matrix, constantly subjected to external and internal stimuli. To this regard, perfused vascularized models are a novel field of investigation that can offer a new technological approach to overcome the limitations of traditional cell culture methods. It allows the combination of perfusion, mechanical and biochemical stimuli, biological cues, biomaterials (mimicking the extracellular matrix of bone), and multiple cell types. This review will discuss macro, milli, and microscale perfused devices designed to model bone structure and microenvironment, focusing on the role of perfusion and encompassing different degrees of complexity. These devices are a very first, though promising, step for the development of 3D in vitro platforms for preclinical screening of novel anabolic or anti-catabolic therapeutic approaches to improve bone health.
Collapse
Affiliation(s)
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Biomedical Science and Technologies Lab, IRCSS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gabriela Graziani
- Laboratory for NanoBiotechnology (NaBi), IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
29
|
Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol 2022; 18:217-231. [PMID: 35058618 DOI: 10.1038/s41584-021-00736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
|
30
|
Atif AR, La̅cis U, Engqvist H, Tenje M, Bagheri S, Mestres G. Experimental Characterization and Mathematical Modeling of the Adsorption of Proteins and Cells on Biomimetic Hydroxyapatite. ACS OMEGA 2022; 7:908-920. [PMID: 35036755 PMCID: PMC8757448 DOI: 10.1021/acsomega.1c05540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Biomaterial development is a long process consisting of multiple stages of design and evaluation within the context of both in vitro and in vivo testing. To streamline this process, mathematical and computational modeling displays potential as a tool for rapid biomaterial characterization, enabling the prediction of optimal physicochemical parameters. In this work, a Langmuir isotherm-based model was used to describe protein and cell adhesion on a biomimetic hydroxyapatite surface, both independently and in a one-way coupled system. The results indicated that increased protein surface coverage leads to improved cell adhesion and spread, with maximal protein coverage occurring within 48 h. In addition, the Langmuir model displayed a good fit with the experimental data. Overall, computational modeling is an exciting avenue that may lead to savings in terms of time and cost during the biomaterial development process.
Collapse
Affiliation(s)
- Abdul-Raouf Atif
- Department
of Materials Science and Engineering, Uppsala
University, Box 35, 751 22 Uppsala, Sweden
| | - Uǵis La̅cis
- Department
of Engineering Mechanics, FLOW Centre, KTH
Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Håkan Engqvist
- Department
of Materials Science and Engineering, Uppsala
University, Box 35, 751 22 Uppsala, Sweden
| | - Maria Tenje
- Department
of Materials Science and Engineering, Uppsala
University, Box 35, 751 22 Uppsala, Sweden
- Science
for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Shervin Bagheri
- Department
of Engineering Mechanics, FLOW Centre, KTH
Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Gemma Mestres
- Department
of Materials Science and Engineering, Uppsala
University, Box 35, 751 22 Uppsala, Sweden
- Science
for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| |
Collapse
|
31
|
Zhang P, Shao N, Qin L. Recent Advances in Microfluidic Platforms for Programming Cell-Based Living Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005944. [PMID: 34270839 DOI: 10.1002/adma.202005944] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/20/2020] [Indexed: 06/13/2023]
Abstract
Cell-based living materials, including single cells, cell-laden fibers, cell sheets, organoids, and organs, have attracted intensive interests owing to their widespread applications in cancer therapy, regenerative medicine, drug development, and so on. Significant progress in materials, microfabrication, and cell biology have promoted the development of numerous promising microfluidic platforms for programming these cell-based living materials with a high-throughput, scalable, and efficient manner. In this review, the recent progress of novel microfluidic platforms for programming cell-based living materials is presented. First, the unique features, categories, and materials and related fabrication methods of microfluidic platforms are briefly introduced. From the viewpoint of the design principles of the microfluidic platforms, the recent significant advances of programming single cells, cell-laden fibers, cell sheets, organoids, and organs in turns are then highlighted. Last, by providing personal perspectives on challenges and future trends, this review aims to motivate researchers from the fields of materials and engineering to work together with biologists and physicians to promote the development of cell-based living materials for human healthcare-related applications.
Collapse
Affiliation(s)
- Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Ning Shao
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| |
Collapse
|
32
|
Vunjak-Novakovic G, Ronaldson-Bouchard K, Radisic M. Organs-on-a-chip models for biological research. Cell 2021; 184:4597-4611. [PMID: 34478657 PMCID: PMC8417425 DOI: 10.1016/j.cell.2021.08.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
We explore the utility of bioengineered human tissues-individually or connected into physiological units-for biological research. While much smaller and simpler than their native counterparts, these tissues are complex enough to approximate distinct tissue phenotypes: molecular, structural, and functional. Unlike organoids, which form spontaneously and recapitulate development, "organs-on-a-chip" are engineered to display some specific functions of whole organs. Looking back, we discuss the key developments of this emerging technology. Thinking forward, we focus on the challenges faced to fully establish, validate, and utilize the fidelity of these models for biological research.
Collapse
|
33
|
Slay EE, Meldrum FC, Pensabene V, Amer MH. Embracing Mechanobiology in Next Generation Organ-On-A-Chip Models of Bone Metastasis. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:722501. [PMID: 35047952 PMCID: PMC8757701 DOI: 10.3389/fmedt.2021.722501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
Bone metastasis in breast cancer is associated with high mortality. Biomechanical cues presented by the extracellular matrix play a vital role in driving cancer metastasis. The lack of in vitro models that recapitulate the mechanical aspects of the in vivo microenvironment hinders the development of novel targeted therapies. Organ-on-a-chip (OOAC) platforms have recently emerged as a new generation of in vitro models that can mimic cell-cell interactions, enable control over fluid flow and allow the introduction of mechanical cues. Biomaterials used within OOAC platforms can determine the physical microenvironment that cells reside in and affect their behavior, adhesion, and localization. Refining the design of OOAC platforms to recreate microenvironmental regulation of metastasis and probe cell-matrix interactions will advance our understanding of breast cancer metastasis and support the development of next-generation metastasis-on-a-chip platforms. In this mini-review, we discuss the role of mechanobiology on the behavior of breast cancer and bone-residing cells, summarize the current capabilities of OOAC platforms for modeling breast cancer metastasis to bone, and highlight design opportunities offered by the incorporation of mechanobiological cues in these platforms.
Collapse
Affiliation(s)
- Ellen E. Slay
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Virginia Pensabene
- School of School of Electronic and Electrical Engineering, University of Leeds, Leeds, United Kingdom
- School of Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
34
|
Li T, Liu Y, Zhang Q, Sun W, Dong Y. A steroid-induced osteonecrosis model established using an organ-on-a-chip platform. Exp Ther Med 2021; 22:1070. [PMID: 34447463 PMCID: PMC8355687 DOI: 10.3892/etm.2021.10504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
Bone microvascular endothelial cells (BMECs) constitute the central part of the femoral head's intramural microenvironment network and have an essential role in the development of steroid-induced osteonecrosis of the femoral head. Recently, the rapid development of microfluidic technology has led to innovations in the fields of chemistry, medicine and life sciences. It is now possible to use microfluidics organ-on-a-chip techniques to assess osteonecrosis. In the present study, BMECs were cultured on a microfluidic organ-on-a-chip platform to explore the pathogenesis of femoral-head necrosis. The aim of the present study was to explore the effects of different interventions on BMECs and study the pathogenesis of steroid-induced osteonecrosis through a microfluidic organ-on-a-chip platform. Methods including SU-8 lithography were used to produce a microfluidic organ-on-a-chip and human umbilical vein endothelial cells (HUVECs) were used to test whether it was possible to culture cells on the chip. Subsequently, a set of methods were applied for the isolation, purification, culture and identification of BMECs. Hydroxyapatite (HA) was used for co-culture, dexamethasone was used at different concentrations as an intervention in the cells and icariin was used for protection. BMECs were isolated and cultured from the femoral head obtained following total hip arthroplasty and were then inoculated into the microfluidic organ-on-a-chip for further treatment. In part I of the experiment, HUVECs and BMECs both successfully survived on the chip and a comparison of the growth and morphology was performed. HA and BMECs were then co-cultured for comparison with the control group. The cell growth was observed by confocal microscopy after 24 h. In part II, the effects of different concentrations of glucocorticoid (0.4 or 0.6 mg/ml dexamethasone) and the protection of icariin were evaluated. The morphology of BMECs and the cleaved caspase-3/7 content were observed by immunofluorescence staining and confocal microscopy after 24 h. In the microfluidic organ-on-a-chip, the response of the cells was able to be accurately observed. In part I, at the same concentration of injected cells, BMECs exhibited improved viability compared with HUVECs (P<0.05). In addition, it was indicated that HA was not only able to promote the germination and growth of BMECs but also improve the survival of the cells (P<0.05). In part II, it was identified that dexamethasone was able to induce BMECs to produce cleaved caspase 3/7; the caspase 3/7 content was significantly higher than that in the blank control group (P<0.05) and a dose correlation was observed. Icariin was able to inhibit this process and protect the microvascular structure of BMECs. The content of cleaved caspase 3/7 in the icariin-protected group was significantly lower than that in the group without icariin (P<0.05). It was concluded that BMECs are more likely to survive than HUVECs and HA promoted the growth of BMECs on the microfluidic organ-on-a-chip platform. Glucocorticoid caused damage to BMECs through the production of cleaved caspase 3/7, which was observed through the microfluidic organ-on-a-chip platform, and icariin protected BMECs from damage.
Collapse
Affiliation(s)
- Tengqi Li
- Department of Orthopedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, P.R. China
| | - Yadi Liu
- Department of Bioanalysis, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Qingyu Zhang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Wei Sun
- Department of Orthopedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, P.R. China.,Department of Orthopedic Surgery, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Yiyang Dong
- Department of Bioanalysis, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| |
Collapse
|
35
|
Larsen JB, Taebnia N, Dolatshahi-Pirouz A, Eriksen AZ, Hjørringgaard C, Kristensen K, Larsen NW, Larsen NB, Marie R, Mündler AK, Parhamifar L, Urquhart AJ, Weller A, Mortensen KI, Flyvbjerg H, Andresen TL. Imaging therapeutic peptide transport across intestinal barriers. RSC Chem Biol 2021; 2:1115-1143. [PMID: 34458827 PMCID: PMC8341777 DOI: 10.1039/d1cb00024a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Oral delivery is a highly preferred method for drug administration due to high patient compliance. However, oral administration is intrinsically challenging for pharmacologically interesting drug classes, in particular pharmaceutical peptides, due to the biological barriers associated with the gastrointestinal tract. In this review, we start by summarizing the pharmacological performance of several clinically relevant orally administrated therapeutic peptides, highlighting their low bioavailabilities. Thus, there is a strong need to increase the transport of peptide drugs across the intestinal barrier to realize future treatment needs and further development in the field. Currently, progress is hampered by a lack of understanding of transport mechanisms that govern intestinal absorption and transport of peptide drugs, including the effects of the permeability enhancers commonly used to mediate uptake. We describe how, for the past decades, mechanistic insights have predominantly been gained using functional assays with end-point read-out capabilities, which only allow indirect study of peptide transport mechanisms. We then focus on fluorescence imaging that, on the other hand, provides opportunities to directly visualize and thus follow peptide transport at high spatiotemporal resolution. Consequently, it may provide new and detailed mechanistic understanding of the interplay between the physicochemical properties of peptides and cellular processes; an interplay that determines the efficiency of transport. We review current methodology and state of the art in the field of fluorescence imaging to study intestinal barrier transport of peptides, and provide a comprehensive overview of the imaging-compatible in vitro, ex vivo, and in vivo platforms that currently are being developed to accelerate this emerging field of research.
Collapse
Affiliation(s)
- Jannik Bruun Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Nayere Taebnia
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Alireza Dolatshahi-Pirouz
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Anne Zebitz Eriksen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Claudia Hjørringgaard
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Kasper Kristensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Nanna Wichmann Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Niels Bent Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Rodolphe Marie
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Ann-Kathrin Mündler
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Ladan Parhamifar
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Andrew James Urquhart
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Arjen Weller
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Kim I Mortensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Henrik Flyvbjerg
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Thomas Lars Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| |
Collapse
|
36
|
Abdul Sisak MA, Louis F, Aoki I, Lee SH, Chang YT, Matsusaki M. A Near-Infrared Organic Fluorescent Probe for Broad Applications for Blood Vessels Imaging by High-Throughput Screening via 3D-Blood Vessel Models. SMALL METHODS 2021; 5:e2100338. [PMID: 34927878 DOI: 10.1002/smtd.202100338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/24/2021] [Indexed: 06/14/2023]
Abstract
Blood vessels are present in all of the organs, reflecting their importance for oxygen and nutrient delivery to the cells. Until now, no organic fluorophore has been reported for the live imaging of endothelium although the layer is the key to blood vessel functions. Here, the discovery of a blood vessel organic probe at near-infrared (NIR) wavelength range (BV-NIR) through an engineered blood capillary-based screening system, which is a more physiological model than a conventional cell culture condition, is reported. This selected Cy5 based probe shows the highest specific adsorption property out of 240 candidates on the endothelium and is equivalent to an anti-CD31 antibody in terms of intensity. The BV-NIR probe indicating strong and stable in vitro, ex vivo, and in vivo imaging of the endothelium even after histological immunostaining processes shows potential as a convenient tool for live imaging as well as for covisualization with a specific antibody.
Collapse
Affiliation(s)
- Muhammad Asri Abdul Sisak
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
| | - Ichio Aoki
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, 263-8555, Japan
| | - Sun Hyeok Lee
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Young-Tae Chang
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, Republic of Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
37
|
Whelan IT, Moeendarbary E, Hoey DA, Kelly DJ. Biofabrication of vasculature in microphysiological models of bone. Biofabrication 2021; 13. [PMID: 34034238 DOI: 10.1088/1758-5090/ac04f7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022]
Abstract
Bone contains a dense network of blood vessels that are essential to its homoeostasis, endocrine function, mineral metabolism and regenerative functions. In addition, bone vasculature is implicated in a number of prominent skeletal diseases, and bone has high affinity for metastatic cancers. Despite vasculature being an integral part of bone physiology and pathophysiology, it is often ignored or oversimplified inin vitrobone models. However, 3D physiologically relevant vasculature can now be engineeredin vitro, with microphysiological systems (MPS) increasingly being used as platforms for engineering this physiologically relevant vasculature. In recent years, vascularised models of bone in MPSs systems have been reported in the literature, representing the beginning of a possible technological step change in how bone is modelledin vitro. Vascularised bone MPSs is a subfield of bone research in its nascency, however given the impact of MPSs has had inin vitroorgan modelling, and the crucial role of vasculature to bone physiology, these systems stand to have a substantial impact on bone research. However, engineering vasculature within the specific design restraints of the bone niche is significantly challenging given the different requirements for engineering bone and vasculature. With this in mind, this paper aims to serve as technical guidance for the biofabrication of vascularised bone tissue within MPS devices. We first discuss the key engineering and biological considerations for engineering more physiologically relevant vasculaturein vitrowithin the specific design constraints of the bone niche. We next explore emerging applications of vascularised bone MPSs, and conclude with a discussion on the current status of vascularised bone MPS biofabrication and suggest directions for development of next generation vascularised bone MPSs.
Collapse
|
38
|
Guttenplan APM, Tahmasebi Birgani Z, Giselbrecht S, Truckenmüller RK, Habibović P. Chips for Biomaterials and Biomaterials for Chips: Recent Advances at the Interface between Microfabrication and Biomaterials Research. Adv Healthc Mater 2021; 10:e2100371. [PMID: 34033239 PMCID: PMC11468311 DOI: 10.1002/adhm.202100371] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/08/2021] [Indexed: 12/24/2022]
Abstract
In recent years, the use of microfabrication techniques has allowed biomaterials studies which were originally carried out at larger length scales to be miniaturized as so-called "on-chip" experiments. These miniaturized experiments have a range of advantages which have led to an increase in their popularity. A range of biomaterial shapes and compositions are synthesized or manufactured on chip. Moreover, chips are developed to investigate specific aspects of interactions between biomaterials and biological systems. Finally, biomaterials are used in microfabricated devices to replicate the physiological microenvironment in studies using so-called "organ-on-chip," "tissue-on-chip" or "disease-on-chip" models, which can reduce the use of animal models with their inherent high cost and ethical issues, and due to the possible use of human cells can increase the translation of research from lab to clinic. This review gives an overview of recent developments at the interface between microfabrication and biomaterials science, and indicates potential future directions that the field may take. In particular, a trend toward increased scale and automation is apparent, allowing both industrial production of micron-scale biomaterials and high-throughput screening of the interaction of diverse materials libraries with cells and bioengineered tissues and organs.
Collapse
Affiliation(s)
- Alexander P. M. Guttenplan
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
39
|
Park S, Gwon Y, Kim W, Kim J. Rebirth of the Eggshell Membrane as a Bioactive Nanoscaffold for Tissue Engineering. ACS Biomater Sci Eng 2021; 7:2219-2224. [PMID: 34061495 DOI: 10.1021/acsbiomaterials.1c00552] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Eggshell membrane (ESM)-based biomaterials have generated significant interest for their potential biomedical applications, including those in tissue engineering and regenerative medicine. Herein, the development of a bioactive ESM-based nanopatterned scaffold for enhancing the adhesion and functions of cells has been described. To control the shape of the raw ESM with entangled protein fibers, a two-step dissolution technique is used. Subsequently, nanoimprint lithography is applied to the ESM solution to fabricate scaffolds with a nanotopographic surface inspired by the fiber alignment of the extracellular matrix. In this way, the morphology and proliferation of attached osteoblasts are sensitively controlled through their response to the nanopatterned topography of the prepared scaffold, allowing significant improvements in their osteogenic differentiation and growth factor secretion. This study demonstrates the potential use of this bioactive ESM-based nanopatterned substrate as an effective cell and tissue engineering scaffold.
Collapse
Affiliation(s)
- Sunho Park
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.,Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yonghyun Gwon
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.,Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Woochan Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.,Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.,Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
40
|
Atif AR, Pujari-Palmer M, Tenje M, Mestres G. A microfluidics-based method for culturing osteoblasts on biomimetic hydroxyapatite. Acta Biomater 2021; 127:327-337. [PMID: 33785452 DOI: 10.1016/j.actbio.2021.03.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/02/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
The reliability of conventional cell culture studies to evaluate biomaterials is often questioned, as in vitro outcomes may contradict results obtained through in vivo assays. Microfluidics technology has the potential to reproduce complex physiological conditions by allowing for fine control of microscale features such as cell confinement and flow rate. Having a continuous flow during cell culture is especially advantageous for bioactive biomaterials such as calcium-deficient hydroxyapatite (HA), which may otherwise alter medium composition and jeopardize cell viability, potentially producing false negative results in vitro. In this work, HA was integrated into a microfluidics-based platform (HA-on-chip) and the effect of varied flow rates (2, 8 and 14 µl/min, corresponding to 0.002, 0.008 and 0.014 dyn/cm2, respectively) was evaluated. A HA sample placed in a well plate (HA-static) was included as a control. While substantial calcium depletion and phosphate release occurred in static conditions, the concentration of ions in HA-on-chip samples remained similar to those of fresh medium, particularly at higher flow rates. Pre-osteoblast-like cells (MC3T3-E1) exhibited a significantly higher degree of proliferation on HA-on-chip (8 μl/min flow rate) as compared to HA-static. However, cell differentiation, analysed by alkaline phosphatase (ALP) activity, showed low values in both conditions. This study indicates that cells respond differently when cultured on HA under flow compared to static conditions, which indicates the need for more physiologically relevant methods to increase the predictive value of in vitro studies used to evaluate biomaterials. STATEMENT OF SIGNIFICANCE: There is a lack of correlation between the results obtained when testing some biomaterials under cell culture as opposed to animal models. To address this issue, a cell culture method with slightly enhanced physiological relevance was developed by incorporating a biomaterial, known to regenerate bone, inside of a microfluidic platform that enabled a continuous supply of cell culture medium. Since the utilized biomaterial interacts with surrounding ions, the perfusion of medium allowed for shielding of these changes similarly as would happen in the body. The experimental outcomes observed in the dynamic platform were different than those obtained with standard static cell culture systems, proving the key role of the platform in the assessment of biomaterials.
Collapse
Affiliation(s)
- Abdul Raouf Atif
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Michael Pujari-Palmer
- Division of Applied Materials Science, Department of Materials Science and Engineering, Uppsala University, 751 22 Uppsala, Sweden
| | - Maria Tenje
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Gemma Mestres
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden.
| |
Collapse
|
41
|
Rial R, González-Durruthy M, Liu Z, Ruso JM. Advanced Materials Based on Nanosized Hydroxyapatite. Molecules 2021; 26:3190. [PMID: 34073479 PMCID: PMC8198166 DOI: 10.3390/molecules26113190] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 02/02/2023] Open
Abstract
The development of new materials based on hydroxyapatite has undergone a great evolution in recent decades due to technological advances and development of computational techniques. The focus of this review is the various attempts to improve new hydroxyapatite-based materials. First, we comment on the most used processing routes, highlighting their advantages and disadvantages. We will now focus on other routes, less common due to their specificity and/or recent development. We also include a block dedicated to the impact of computational techniques in the development of these new systems, including: QSAR, DFT, Finite Elements of Machine Learning. In the following part we focus on the most innovative applications of these materials, ranging from medicine to new disciplines such as catalysis, environment, filtration, or energy. The review concludes with an outlook for possible new research directions.
Collapse
Affiliation(s)
- Ramón Rial
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (R.R.); (M.G.-D.)
| | - Michael González-Durruthy
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (R.R.); (M.G.-D.)
| | - Zhen Liu
- Department of Physics and Engineering, Frostburg State University, Frostburg, MD 21532, USA;
| | - Juan M. Ruso
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (R.R.); (M.G.-D.)
| |
Collapse
|
42
|
Designing Hydrogel-Based Bone-On-Chips for Personalized Medicine. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11104495] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The recent development of bone-on-chips (BOCs) holds the main advantage of requiring a low quantity of cells and material, compared to traditional In Vitro models. By incorporating hydrogels within BOCs, the culture system moved to a three dimensional culture environment for cells which is more representative of bone tissue matrix and function. The fundamental components of hydrogel-based BOCs, namely the cellular sources, the hydrogel and the culture chamber, have been tuned to mimic the hematopoietic niche in the bone aspirate marrow, cancer bone metastasis and osteo/chondrogenic differentiation. In this review, we examine the entire process of developing hydrogel-based BOCs to model In Vitro a patient specific situation. First, we provide bone biological understanding for BOCs design and then how hydrogel structural and mechanical properties can be tuned to meet those requirements. This is followed by a review on hydrogel-based BOCs, developed in the last 10 years, in terms of culture chamber design, hydrogel and cell source used. Finally, we provide guidelines for the definition of personalized pathological and physiological bone microenvironments. This review covers the information on bone, hydrogel and BOC that are required to develop personalized therapies for bone disease, by recreating clinically relevant scenarii in miniaturized devices.
Collapse
|
43
|
Bessy T, Itkin T, Passaro D. Bioengineering the Bone Marrow Vascular Niche. Front Cell Dev Biol 2021; 9:645496. [PMID: 33996805 PMCID: PMC8113773 DOI: 10.3389/fcell.2021.645496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/23/2021] [Indexed: 01/01/2023] Open
Abstract
The bone marrow (BM) tissue is the main physiological site for adult hematopoiesis. In recent years, the cellular and matrix components composing the BM have been defined with unprecedent resolution, both at the molecular and structural levels. With the expansion of this knowledge, the possibility of reproducing a BM-like structure, to ectopically support and study hematopoiesis, becomes a reality. A number of experimental systems have been implemented and have displayed the feasibility of bioengineering BM tissues, supported by cells of mesenchymal origin. Despite being known as an abundant component of the BM, the vasculature has been largely disregarded for its role in regulating tissue formation, organization and determination. Recent reports have highlighted the crucial role for vascular endothelial cells in shaping tissue development and supporting steady state, emergency and malignant hematopoiesis, both pre- and postnatally. Herein, we review the field of BM-tissue bioengineering with a particular focus on vascular system implementation and integration, starting from describing a variety of applicable in vitro models, ending up with in vivo preclinical models. Additionally, we highlight the challenges of the field and discuss the clinical perspectives in terms of adoptive transfer of vascularized BM-niche grafts in patients to support recovering hematopoiesis.
Collapse
Affiliation(s)
- Thomas Bessy
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| | - Tomer Itkin
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Diana Passaro
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
44
|
Lim J, Ching H, Yoon JK, Jeon NL, Kim Y. Microvascularized tumor organoids-on-chips: advancing preclinical drug screening with pathophysiological relevance. NANO CONVERGENCE 2021; 8:12. [PMID: 33846849 PMCID: PMC8042002 DOI: 10.1186/s40580-021-00261-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/17/2021] [Indexed: 05/06/2023]
Abstract
Recent developments of organoids engineering and organ-on-a-chip microfluidic technologies have enabled the recapitulation of the major functions and architectures of microscale human tissue, including tumor pathophysiology. Nevertheless, there remain challenges in recapitulating the complexity and heterogeneity of tumor microenvironment. The integration of these engineering technologies suggests a potential strategy to overcome the limitations in reconstituting the perfusable microvascular system of large-scale tumors conserving their key functional features. Here, we review the recent progress of in vitro tumor-on-a-chip microfluidic technologies, focusing on the reconstruction of microvascularized organoid models to suggest a better platform for personalized cancer medicine.
Collapse
Affiliation(s)
- Jungeun Lim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- George W, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA, 30332, USA
| | - Hanna Ching
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeong-Kee Yoon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Noo Li Jeon
- George W, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA, 30332, USA
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - YongTae Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
45
|
Tronolone JJ, Jain A. Engineering new microvascular networks on-chip: ingredients, assembly, and best practices. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007199. [PMID: 33994903 PMCID: PMC8114943 DOI: 10.1002/adfm.202007199] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 05/23/2023]
Abstract
Tissue engineered grafts show great potential as regenerative implants for diseased or injured tissues within the human body. However, these grafts suffer from poor nutrient perfusion and waste transport, thus decreasing their viability post-transplantation. Graft vascularization is therefore a major area of focus within tissue engineering because biologically relevant conduits for nutrient and oxygen perfusion can improve viability post-implantation. Many researchers utilize microphysiological systems as testing platforms for potential grafts due to an ability to integrate vascular networks as well as biological characteristics such as fluid perfusion, 3D architecture, compartmentalization of tissue-specific materials, and biophysical and biochemical cues. While many methods of vascularizing these systems exist, microvascular self-assembly has great potential for bench-to-clinic translation as it relies on naturally occurring physiological events. In this review, we highlight the past decade of literature and critically discuss the most important and tunable components yielding a self-assembled vascular network on chip: endothelial cell source, tissue-specific supporting cells, biomaterial scaffolds, biochemical cues, and biophysical forces. This article discusses the bioengineered systems of angiogenesis, vasculogenesis, and lymphangiogenesis, and includes a brief overview of multicellular systems. We conclude with future avenues of research to guide the next generation of vascularized microfluidic models and future tissue engineered grafts.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Abhishek Jain
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77808, USA
| |
Collapse
|
46
|
Gwon Y, Park S, Kim W, Han T, Kim H, Kim J. Radially patterned transplantable biodegradable scaffolds as topographically defined contact guidance platforms for accelerating bone regeneration. J Biol Eng 2021; 15:12. [PMID: 33752709 PMCID: PMC7986475 DOI: 10.1186/s13036-021-00263-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The healing of large critical-sized bone defects remains a clinical challenge in modern orthopedic medicine. The current gold standard for treating critical-sized bone defects is autologous bone graft; however, it has critical limitations. Bone tissue engineering has been proposed as a viable alternative, not only for replacing the current standard treatment, but also for producing complete regeneration of bone tissue without complex surgical treatments or tissue transplantation. In this study, we proposed a transplantable radially patterned scaffold for bone regeneration that was defined by capillary force lithography technology using biodegradable polycaprolactone polymer. RESULTS The radially patterned transplantable biodegradable scaffolds had a radial structure aligned in a central direction. The radially aligned pattern significantly promoted the recruitment of host cells and migration of osteoblasts into the defect site. Furthermore, the transplantable scaffolds promoted regeneration of critical-sized bone defects by inducing cell migration and differentiation. CONCLUSIONS Our findings demonstrated that topographically defined radially patterned transplantable biodegradable scaffolds may have great potential for clinical application of bone tissue regeneration.
Collapse
Affiliation(s)
- Yonghyun Gwon
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunho Park
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Woochan Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Taeseong Han
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoseong Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
47
|
Mansoorifar A, Gordon R, Bergan R, Bertassoni LE. Bone-on-a-chip: microfluidic technologies and microphysiologic models of bone tissue. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2006796. [PMID: 35422682 PMCID: PMC9007546 DOI: 10.1002/adfm.202006796] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 05/07/2023]
Abstract
Bone is an active organ that continuously undergoes an orchestrated process of remodeling throughout life. Bone tissue is uniquely capable of adapting to loading, hormonal, and other changes happening in the body, as well as repairing bone that becomes damaged to maintain tissue integrity. On the other hand, diseases such as osteoporosis and metastatic cancers disrupt normal bone homeostasis leading to compromised function. Historically, our ability to investigate processes related to either physiologic or diseased bone tissue has been limited by traditional models that fail to emulate the complexity of native bone. Organ-on-a-chip models are based on technological advances in tissue engineering and microfluidics, enabling the reproduction of key features specific to tissue microenvironments within a microfabricated device. Compared to conventional in-vitro and in-vivo bone models, microfluidic models, and especially organs-on-a-chip platforms, provide more biomimetic tissue culture conditions, with increased predictive power for clinical assays. In this review, we will report microfluidic and organ-on-a-chip technologies designed for understanding the biology of bone as well as bone-related diseases and treatments. Finally, we discuss the limitations of the current models and point toward future directions for microfluidics and organ-on-a-chip technologies in bone research.
Collapse
Affiliation(s)
- Amin Mansoorifar
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Ryan Gordon
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Luiz E. Bertassoni
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, USA
| |
Collapse
|
48
|
Sharma V, Srinivasan A, Nikolajeff F, Kumar S. Biomineralization process in hard tissues: The interaction complexity within protein and inorganic counterparts. Acta Biomater 2021; 120:20-37. [PMID: 32413577 DOI: 10.1016/j.actbio.2020.04.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/17/2020] [Accepted: 04/26/2020] [Indexed: 02/07/2023]
Abstract
Biomineralization can be considered as nature's strategy to produce and sustain biominerals, primarily via creation of hard tissues for protection and support. This review examines the biomineralization process within the hard tissues of the human body with special emphasis on the mechanisms and principles of bone and teeth mineralization. We describe the detailed role of proteins and inorganic ions in mediating the mineralization process. Furthermore, we highlight the various available models for studying bone physiology and mineralization starting from the historical static cell line-based methods to the most advanced 3D culture systems, elucidating the pros and cons of each one of these methods. With respect to the mineralization process in teeth, enamel and dentin mineralization is discussed in detail. The key role of intrinsically disordered proteins in modulating the process of mineralization in enamel and dentine is given attention. Finally, nanotechnological interventions in the area of bone and teeth mineralization, diseases and tissue regeneration is also discussed. STATEMENT OF SIGNIFICANCE: This article provides an overview of the biomineralization process within hard tissues of the human body, which encompasses the detailed mechanism innvolved in the formation of structures like teeth and bone. Moreover, we have discussed various available models used for studying biomineralization and also explored the nanotechnological applications in the field of bone regeneration and dentistry.
Collapse
Affiliation(s)
- Vaibhav Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| | | | | | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
49
|
Fabrication of Pentoxifylline-Loaded Hydroxyapatite/Alginate Scaffold for Bone Tissue Engineering. JOURNAL OF BIOMIMETICS BIOMATERIALS AND BIOMEDICAL ENGINEERING 2020. [DOI: 10.4028/www.scientific.net/jbbbe.47.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background: Hydroxyapatite (HAP), as a common biomaterial in bone tissue engineering, can be fabricated in combination with other osteogenic agents. Pentoxifylline (PTX) is demonstrated to have positive roles in bone defect healing. Since local administration can diminish the systemic side effects of the drug, the objectives of the current in vitro study were to find the effects of PTX on the osteoblast functions for tissue engineering applications. Methods: a HAP scaffold was fabricated by casting the HAP slurry within polyurethane foam. The scaffold was enriched with 5 mg/mL PTX. Alginate (Alg) was used as drug carrier to regulate the PTX releasing rate. MG-63 osteosarcoma cells were cultured on 3D scaffolds and 2D Alg films in the presence or absence of PTX. Results: PTX did not affect the cell viability, attachment and phenotype. Also, the ultrastructure of the scaffolds was not modified by PTX enrichment. Alizarin red S staining showed that PTX has no effect on calcium deposition. Besides, Raman confocal microscopy demonstrated an increase in the organic matrix formation including proline, valine and phenylalanine deposition (represented collagen). Although PTX increased the total protein secretion, it led to a decrease in the alkaline phosphatase activity and vascular endothelial growth factor (VEGF) content. PTX reduced the hydration and degradation rates and it was released mainly at the first 24 hours of incubation. Conclusion: Based on our in vitro study, application of engineered PTX-loaded HAP scaffold in bone regeneration can act on behalf of organic matrix production, but not angiogenesis and mineralization.
Collapse
|
50
|
Peticone C, Thompson DDS, Dimov N, Jevans B, Glass N, Micheletti M, Knowles JC, Kim HW, Cooper-White JJ, Wall IB. Characterisation of osteogenic and vascular responses of hMSCs to Ti-Co doped phosphate glass microspheres using a microfluidic perfusion platform. J Tissue Eng 2020; 11:2041731420954712. [PMID: 33178409 PMCID: PMC7592314 DOI: 10.1177/2041731420954712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/13/2020] [Indexed: 01/22/2023] Open
Abstract
Using microspherical scaffolds as building blocks to repair bone defects of
specific size and shape has been proposed as a tissue engineering strategy.
Here, phosphate glass (PG) microcarriers doped with 5 mol % TiO2 and
either 0 mol % CoO (CoO 0%) or 2 mol % CoO (CoO 2%) were investigated for their
ability to support osteogenic and vascular responses of human mesenchymal stem
cells (hMSCs). Together with standard culture techniques, cell-material
interactions were studied using a novel perfusion microfluidic bioreactor that
enabled cell culture on microspheres, along with automated processing and
screening of culture variables. While titanium doping was found to support hMSCs
expansion and differentiation, as well as endothelial cell-derived vessel
formation, additional doping with cobalt did not improve the functionality of
the microspheres. Furthermore, the microfluidic bioreactor enabled screening of
culture parameters for cell culture on microspheres that could be potentially
translated to a scaled-up system for tissue-engineered bone manufacturing.
Collapse
Affiliation(s)
- Carlotta Peticone
- Department of Biochemical Engineering, University College London, London, UK
| | | | - Nikolay Dimov
- Centre for Engineering Research, University of Hertfordshire, Hatfield, Hertfordshire, UK
| | - Ben Jevans
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Nick Glass
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Brisbane, Australia
| | - Martina Micheletti
- Department of Biochemical Engineering, University College London, London, UK
| | - Jonathan C Knowles
- Division of Biomaterials and Tissue Engineering, University College London Eastman Dental Institute, London, UK.,The Discoveries Centre for Regenerative and Precision Medicine, UCL Campus, London, UK.,Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute for Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
| | - Hae-Won Kim
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute for Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
| | - Justin J Cooper-White
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Brisbane, Australia.,School of Chemical Engineering, University of Queensland, St. Lucia, Brisbane, Australia
| | - Ivan B Wall
- Department of Biochemical Engineering, University College London, London, UK.,Institute for Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea.,Aston Medical Research Institute and School of Life and Health Sciences, Aston University, Birmingham, UK
| |
Collapse
|