1
|
Roehm KD, Chiesa I, Haithcock D, Gottardi R, Prabhakarpandian B. A vascularized microfluidic model of the osteochondral unit for modeling inflammatory response and therapeutic screening. LAB ON A CHIP 2024. [PMID: 39715348 DOI: 10.1039/d4lc00651h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Osteoarthritis (OA) has long been considered a disease of the articular cartilage. Within the past decade it has become increasingly clear that OA is a disease of the entire joint space and that interactions between articular cartilage and subchondral bone likely play an important role in the disease. Driven by this knowledge, we have created a novel microphysiological model of the osteochondral unit containing synovium, cartilage, bone, and vasculature in separate compartments with molecular and direct cell-cell interaction between the cells from the different tissue types. We have characterized the model in terms of differentiation by molecule and matrix secretion and shown that it demonstrates morphology and functionality that mimic the native characteristic of the joint space. Finally, we induced inflammation and subsequently rescued the model constructs by a known compound as proof of concept for anti-inflammatory drug screening applications.
Collapse
Affiliation(s)
- Kevin D Roehm
- CFD Research Corporation, 6820 Moquin Dr. N.W., Huntsville, AL 35806, USA.
| | - Irene Chiesa
- Department of Information Engineering and Research Center "Enrico Piaggio", University of Pisa, Italy
- Division of Otolaryngology, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dustin Haithcock
- CFD Research Corporation, 6820 Moquin Dr. N.W., Huntsville, AL 35806, USA.
| | - Riccardo Gottardi
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Division of Otolaryngology, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Pulmonary Medicine, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Ri.MED Foundation, Palermo, Italy
| | | |
Collapse
|
2
|
Adams M, Cottrell J. Development and characterization of an in vitro fluorescently tagged 3D bone-cartilage interface model. Front Endocrinol (Lausanne) 2024; 15:1484912. [PMID: 39600948 PMCID: PMC11588493 DOI: 10.3389/fendo.2024.1484912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Three-dimensional cultures are widely used to study bone and cartilage. These models often focus on the interaction between osteoblasts and osteoclasts or osteoblasts and chondrocytes. A culture of osteoblasts, osteoclasts and chondrocytes would represent the cells that interact in the joint and a model with these cells could be used to study many diseases that affect the joints. The goal of this study was to develop 3D bone-cartilage interface (3D-BCI) that included osteoblasts, osteocytes, osteoclasts, and cartilage. Fluorescently tagged cell lines were developed to assess the interactions as cells differentiate to form bone and cartilage. Mouse cell line, MC3T3, was labeled with a nuclear GFP tag and differentiated into osteoblasts and osteocytes in Matrigel. Raw264.7 cells transfected with a red cytoplasmic tag were added to the system and differentiated with the MC3T3 cells to form osteoclasts. A new method was developed to differentiate chondrocyte cell line ATDC5 in a cartilage spheroid, and the ATDC5 spheroid was added to the MC3T3 and Raw264.7 cell model. We used an Incucyte and functional analysis to assess the cells throughout the differentiation process. The 3D-BCI model was found to be positive for TRAP, ALP, Alizarin red and Alcian blue staining to confirm osteoblastogenesis, osteoclastogenesis, and cartilage formation. Gene expression confirmed differentiation of cells based on increased expression of osteoblast markers: Alpl, Bglap, Col1A2, and Runx2, cartilage markers: Acan, Col2A1, Plod2, and osteoclast markers: Acp5, Rank and Ctsk. Based on staining, protein expression and gene expression results, we conclude that we successfully developed a mouse model with a 3D bone-cartilage interface.
Collapse
Affiliation(s)
- Mary Adams
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, United States
- Immunology Translational Research, Translational Early Development, Bristol Myers Squibb, Summit, NJ, United States
| | - Jessica Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, United States
| |
Collapse
|
3
|
Mishra A, Kumar R, Harilal S, Nigam M, Datta D, Singh S. Emerging Landscape of In Vitro Models for Assessing Rheumatoid Arthritis Management. ACS Pharmacol Transl Sci 2024; 7:2280-2305. [PMID: 39144547 PMCID: PMC11320735 DOI: 10.1021/acsptsci.4c00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024]
Abstract
Rheumatoid arthritis (RA) is a complex condition that is influenced by various causes, including immunological, genetic, and environmental factors. Several studies using animal models have documented immune system dysfunction and described the clinical characteristics of the disease. These studies have provided valuable insights into the pathogenesis of inflammatory arthritis and the identification of new targets for treatment. Nevertheless, none of these animal models successfully replicated all the characteristics of RA. Additionally, numerous experimental medications, which were developed based on our enhanced comprehension of the immune system's function in RA, have shown potential in animal research but ultimately proved ineffective during different stages of clinical trials. There have been several novel therapy alternatives, which do not achieve a consistently outstanding therapeutic outcome in all patients. This underscores the importance of employing the progress in in vitro models, particularly 3D models like tissue explants, and diverse multicomponent approaches such as coculture strategies, synovial membrane, articular cartilage, and subchondral bone models that accurately replicate the structural characteristics of RA pathophysiology. These methods are crucial for the advancement of potential therapeutic strategies. This review discusses the latest advancements in in vitro models and their potential to greatly impact research on managing RA.
Collapse
Affiliation(s)
- Abhay
Prakash Mishra
- Department
of Pharmacology, University of Free State, Bloemfontein 9301, South Africa
- Department
of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Rajesh Kumar
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Seetha Harilal
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Manisha Nigam
- Department
of Biochemistry, Hemvati Nandan Bahuguna
Garhwal University, Srinagar
Garhwal, Uttarakhand 246174, India
| | - Deepanjan Datta
- Department
of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sudarshan Singh
- Office of
Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
- Faculty of
Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Primorac D, Molnar V, Tsoukas D, Uzieliene I, Tremolada C, Brlek P, Klarić E, Vidović D, Zekušić M, Pachaleva J, Bernotiene E, Wilson A, Mobasheri A. Tissue engineering and future directions in regenerative medicine for knee cartilage repair: a comprehensive review. Croat Med J 2024; 65:268-287. [PMID: 38868973 PMCID: PMC11157252 DOI: 10.3325/cmj.2024.65.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 01/06/2025] Open
Abstract
This review evaluates the current landscape and future directions of regenerative medicine for knee cartilage repair, with a particular focus on tissue engineering strategies. In this context, scaffold-based approaches have emerged as promising solutions for cartilage regeneration. Synthetic scaffolds, while offering superior mechanical properties, often lack the biological cues necessary for effective tissue integration. Natural scaffolds, though biocompatible and biodegradable, frequently suffer from inadequate mechanical strength. Hybrid scaffolds, combining elements of both synthetic and natural materials, present a balanced approach, enhancing both mechanical support and biological functionality. Advances in decellularized extracellular matrix scaffolds have shown potential in promoting cell infiltration and integration with native tissues. Additionally, bioprinting technologies have enabled the creation of complex, bioactive scaffolds that closely mimic the zonal organization of native cartilage, providing an optimal environment for cell growth and differentiation. The review also explores the potential of gene therapy and gene editing techniques, including CRISPR-Cas9, to enhance cartilage repair by targeting specific genetic pathways involved in tissue regeneration. The integration of these advanced therapies with tissue engineering approaches holds promise for developing personalized and durable treatments for knee cartilage injuries and osteoarthritis. In conclusion, this review underscores the importance of continued multidisciplinary collaboration to advance these innovative therapies from bench to bedside and improve outcomes for patients with knee cartilage damage.
Collapse
Affiliation(s)
- Dragan Primorac
- Dragan Primorac, Poliklinika Sv. Katarina, Branimirova 71E, 10000 Zagreb, Croatia,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Petta D, D'Arrigo D, Salehi S, Talò G, Bonetti L, Vanoni M, Deabate L, De Nardo L, Dubini G, Candrian C, Moretti M, Lopa S, Arrigoni C. A personalized osteoarthritic joint-on-a-chip as a screening platform for biological treatments. Mater Today Bio 2024; 26:101072. [PMID: 38757057 PMCID: PMC11097088 DOI: 10.1016/j.mtbio.2024.101072] [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: 11/29/2023] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Osteoarthritis (OA) is a highly disabling pathology, characterized by synovial inflammation and cartilage degeneration. Orthobiologics have shown promising results in OA treatment thanks to their ability to influence articular cells and modulate the inflammatory OA environment. Considering their complex mechanism of action, the development of reliable and relevant joint models appears as crucial to select the best orthobiologics for each patient. The aim of this study was to establish a microfluidic OA model to test therapies in a personalized human setting. The joint-on-a-chip model included cartilage and synovial compartments, containing hydrogel-embedded chondrocytes and synovial fibroblasts, separated by a channel for synovial fluid. For the cartilage compartment, a Hyaluronic Acid-based matrix was selected to preserve chondrocyte phenotype. Adding OA synovial fluid induced the production of inflammatory cytokines and degradative enzymes, generating an OA microenvironment. Personalized models were generated using patient-matched cells and synovial fluid to test the efficacy of mesenchymal stem cells on OA signatures. The patient-specific models allowed monitoring changes induced by cell injection, highlighting different individual responses to the treatment. Altogether, these results support the use of this joint-on-a-chip model as a prognostic tool to screen the patient-specific efficacy of orthobiologics.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
| | - Daniele D'Arrigo
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- ISBE-SYSBIO Centre of Systems Biology, Milan, Italy at Department of Biotechnology and Biosciences, Università Degli Studi di Milano Bicocca, Piazza Della Scienza 2, 20126, Milan, Italy
| | - Shima Salehi
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Marco Vanoni
- ISBE-SYSBIO Centre of Systems Biology, Milan, Italy at Department of Biotechnology and Biosciences, Università Degli Studi di Milano Bicocca, Piazza Della Scienza 2, 20126, Milan, Italy
| | - Luca Deabate
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Gabriele Dubini
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Christian Candrian
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| |
Collapse
|
6
|
Locke RC, Zlotnick HM, Stoeckl BD, Fryhofer GW, Galarraga JH, Dhand AP, Zgonis MH, Carey JL, Burdick JA, Mauck RL. Linguistic Analysis Identifies Emergent Biomaterial Fabrication Trends for Orthopaedic Applications. Adv Healthc Mater 2023; 12:e2202591. [PMID: 36657736 PMCID: PMC10121863 DOI: 10.1002/adhm.202202591] [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: 10/09/2022] [Revised: 12/20/2022] [Indexed: 01/21/2023]
Abstract
The expanse of publications in tissue engineering (TE) and orthopedic TE (OTE) over the past 20 years presents an opportunity to probe emergent trends in the field to better guide future technologies that can make an impact on musculoskeletal therapies. Leveraging this trove of knowledge, a hierarchical systematic search method and trend analysis using connected network mapping of key terms is developed. Within discrete time intervals, an accelerated publication rate for anatomic orthopedic tissue engineering (AOTE) of osteochondral defects, tendons, menisci, and entheses is identified. Within these growing fields, the top-listed key terms are extracted and stratified into evident categories, such as biomaterials, delivery method, or 3D printing and biofabrication. It is then identified which categories decreased, remained constant, increased, or emerged over time, identifying the specific emergent categories currently driving innovation in orthopedic repair technologies. Together, these data demonstrate a significant convergence of material types and descriptors used across tissue types. From this convergence, design criteria to support future research of anatomic constructs that mimic both the form and function of native tissues are formulated. In summary, this review identifies large-scale trends and predicts new directions in orthopedics that will define future materials and technologies.
Collapse
Affiliation(s)
- Ryan C. Locke
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Veterans Affairs, CMCVAMC, Philadelphia, PA, USA
| | - Hannah M. Zlotnick
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Veterans Affairs, CMCVAMC, Philadelphia, PA, USA
| | - Brendan D. Stoeckl
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Veterans Affairs, CMCVAMC, Philadelphia, PA, USA
| | - George W. Fryhofer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Abhishek P. Dhand
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Miltiadis H. Zgonis
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - James L. Carey
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason A. Burdick
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Veterans Affairs, CMCVAMC, Philadelphia, PA, USA
| |
Collapse
|
7
|
Monteiro RF, Bakht SM, Gomez-Florit M, Stievani FC, Alves ALG, Reis RL, Gomes ME, Domingues RMA. Writing 3D In Vitro Models of Human Tendon within a Biomimetic Fibrillar Support Platform. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 36952613 DOI: 10.1021/acsami.2c22371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Tendinopathies are poorly understood diseases for which treatment remains challenging. Relevant in vitro models to study human tendon physiology and pathophysiology are therefore highly needed. Here we propose the automated 3D writing of tendon microphysiological systems (MPSs) embedded in a biomimetic fibrillar support platform based on cellulose nanocrystals (CNCs) self-assembly. Tendon decellularized extracellular matrix (dECM) was used to formulate bioinks that closely recapitulate the biochemical signature of tendon niche. A monoculture system recreating the cellular patterns and phenotype of the tendon core was first developed and characterized. This system was then incorporated with a vascular compartment to study the crosstalk between the two cell populations. The combined biophysical and biochemical cues of the printed pattern and dECM hydrogel were revealed to be effective in inducing human-adipose-derived stem cells (hASCs) differentiation toward the tenogenic lineage. In the multicellular system, chemotactic effects promoted endothelial cells migration toward the direction of the tendon core compartment, while the established cellular crosstalk boosted hASCs tenogenesis, emulating the tendon development stages. Overall, the proposed concept is a promising strategy for the automated fabrication of humanized organotypic tendon-on-chip models that will be a valuable new tool for the study of tendon physiology and pathogenesis mechanisms and for testing new tendinopathy treatments.
Collapse
Affiliation(s)
- Rosa F Monteiro
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4800 Braga/Guimarães, Portugal
| | - Syeda M Bakht
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4800 Braga/Guimarães, Portugal
| | - Manuel Gomez-Florit
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4800 Braga/Guimarães, Portugal
| | - Fernanda C Stievani
- Department of Veterinary Surgery and Animal Reproduction, Regenerative Medicine Laboratory, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18607-400 Botucatu, Brazil
| | - Ana L G Alves
- Department of Veterinary Surgery and Animal Reproduction, Regenerative Medicine Laboratory, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18607-400 Botucatu, Brazil
| | - Rui L Reis
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4800 Braga/Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4800 Braga/Guimarães, Portugal
| | - Rui M A Domingues
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4800 Braga/Guimarães, Portugal
| |
Collapse
|
8
|
Stoddart MJ, Della Bella E, Armiento AR. Cartilage Tissue Engineering: An Introduction. Methods Mol Biol 2023; 2598:1-7. [PMID: 36355280 DOI: 10.1007/978-1-0716-2839-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Once damaged, cartilage has limited healing capability. This has led to a huge body of research that aims to repair or regenerate this important tissue. Despite the progress made, significant hurdles still need to be overcome. This chapter highlights some of the progress made, while elaborating on areas that need further research. The concept of translation and the route to clinical translation must be kept in mind if some of the promising preclinical research is to make it to routine clinical application.
Collapse
Affiliation(s)
| | | | - Angela R Armiento
- AO Research Institute Davos, Davos Platz, Switzerland
- UCB Pharma, Slough, UK
| |
Collapse
|
9
|
Galla R, Ruga S, Ferrari S, Saccone S, Saccuman L, Invernizzi M, Uberti F. In vitro analysis of the effects of plant-derived chondroitin sulfate from intestinal barrier to chondrocytes. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
10
|
Badillo-Mata JA, Camacho-Villegas TA, Lugo-Fabres PH. 3D Cell Culture as Tools to Characterize Rheumatoid Arthritis Signaling and Development of New Treatments. Cells 2022; 11:3410. [PMID: 36359806 PMCID: PMC9656230 DOI: 10.3390/cells11213410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 08/08/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune disorders affecting 0.5-1% of the population worldwide. As a disease of multifactorial etiology, its constant study has made it possible to unravel the pathophysiological processes that cause the illness. However, efficient and validated disease models are necessary to continue the search for new disease-modulating drugs. Technologies, such as 3D cell culture and organ-on-a-chip, have contributed to accelerating the prospecting of new therapeutic molecules and even helping to elucidate hitherto unknown aspects of the pathogenesis of multiple diseases. These technologies, where medicine and biotechnology converge, can be applied to understand RA. This review discusses the critical elements of RA pathophysiology and current treatment strategies. Next, we discuss 3D cell culture and apply these methodologies for rheumatological diseases and selected models for RA. Finally, we summarize the application of 3D cell culture for RA treatment.
Collapse
Affiliation(s)
- Jessica Andrea Badillo-Mata
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), A.C. Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| | - Tanya Amanda Camacho-Villegas
- CONACYT-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), A.C. Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| | - Pavel Hayl Lugo-Fabres
- CONACYT-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), A.C. Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
11
|
Dwivedi G, Flaman L, Alaybeyoglu B, Struglics A, Frank EH, Chubinskya S, Trippel SB, Rosen V, Cirit M, Grodzinsky AJ. Inflammatory cytokines and mechanical injury induce post-traumatic osteoarthritis-like changes in a human cartilage-bone-synovium microphysiological system. Arthritis Res Ther 2022; 24:198. [PMID: 35982461 PMCID: PMC9386988 DOI: 10.1186/s13075-022-02881-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/23/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Traumatic knee injuries in humans trigger an immediate increase in synovial fluid levels of inflammatory cytokines that accompany impact damage to joint tissues. We developed a human in vitro cartilage-bone-synovium (CBS) coculture model to study the role of mechanical injury and inflammation in the initiation of post-traumatic osteoarthritis (PTOA)-like disease. METHODS Osteochondral plugs (cartilage-bone, CB) along with joint capsule synovium explants (S) were harvested from 25 cadaveric distal femurs from 16 human donors (Collin's grade 0-2, 23-83years). Two-week monocultures (cartilage (C), bone (B), synovium (S)) and cocultures (CB, CBS) were established. A PTOA-like disease group was initiated via coculture of synovium explants with mechanically impacted osteochondral plugs (CBS+INJ, peak stress 5MPa) with non-impacted CB as controls. Disease-like progression was assessed through analyses of changes in cell viability, inflammatory cytokines released to media (10-plex ELISA), tissue matrix degradation, and metabolomics profile. RESULTS Immediate increases in concentrations of a panel of inflammatory cytokines occurred in CBS+INJ and CBS cocultures and cultures with S alone (IL-1, IL-6, IL-8, and TNF-α among others). CBS+INJ and CBS also showed increased chondrocyte death compared to uninjured CB. The release of sulfated glycosaminoglycans (sGAG) and associated ARGS-aggrecan neoepitope fragments to the medium was significantly increased in CBS and CBS+INJ groups. Distinct metabolomics profiles were observed for C, B, and S monocultures, and metabolites related to inflammatory response in CBS versus CB (e.g., kynurenine, 1-methylnicotinamide, and hypoxanthine) were identified. CONCLUSION CBS and CBS+INJ models showed distinct cellular, inflammatory, and matrix-related alterations relevant to PTOA-like initiation/progression. The use of human knee tissues from donors that had no prior history of OA disease suggests the relevance of this model in highlighting the role of injury and inflammation in earliest stages of PTOA progression.
Collapse
Affiliation(s)
- Garima Dwivedi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA USA
| | - Lisa Flaman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Begum Alaybeyoglu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Javelin Biotech, Woburn, MA USA
| | - André Struglics
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Eliot H. Frank
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Susan Chubinskya
- Departments of Pediatrics, Orthopedic Surgery and Medicine (Section of Rheumatology), Rush University Medical Center, Chicago, IL USA
| | - Stephen B. Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA USA
| | | | - Alan J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, NE47-377, 500 Technology Square, Cambridge, MA 02139 USA
| |
Collapse
|
12
|
Li Z, Lin Z, Liu S, Yagi H, Zhang X, Yocum L, Romero‐Lopez M, Rhee C, Makarcyzk MJ, Yu I, Li EN, Fritch MR, Gao Q, Goh KB, O'Donnell B, Hao T, Alexander PG, Mahadik B, Fisher JP, Goodman SB, Bunnell BA, Tuan RS, Lin H. Human Mesenchymal Stem Cell-Derived Miniature Joint System for Disease Modeling and Drug Testing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105909. [PMID: 35436042 PMCID: PMC9313499 DOI: 10.1002/advs.202105909] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/04/2022] [Indexed: 05/12/2023]
Abstract
Diseases of the knee joint such as osteoarthritis (OA) affect all joint elements. An in vitro human cell-derived microphysiological system capable of simulating intraarticular tissue crosstalk is desirable for studying etiologies/pathogenesis of joint diseases and testing potential therapeutics. Herein, a human mesenchymal stem cell-derived miniature joint system (miniJoint) is generated, in which engineered osteochondral complex, synovial-like fibrous tissue, and adipose tissue are integrated into a microfluidics-enabled bioreactor. This novel design facilitates different tissues communicating while still maintaining their respective phenotypes. The miniJoint exhibits physiologically relevant changes when exposed to interleukin-1β mediated inflammation, which are similar to observations in joint diseases in humans. The potential of the miniJoint in predicting in vivo efficacy of drug treatment is confirmed by testing the "therapeutic effect" of the nonsteroidal anti-inflammatory drug, naproxen, as well as four other potential disease-modifying OA drugs. The data demonstrate that the miniJoint recapitulates complex tissue interactions, thus providing a robust organ chip model for the study of joint pathology and the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Zhong Li
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Zixuan Lin
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Silvia Liu
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPA15261USA
| | - Haruyo Yagi
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Xiurui Zhang
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Lauren Yocum
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | | | - Claire Rhee
- Department of Orthopaedic SurgeryStanford UniversityStanfordCA94305USA
| | - Meagan J. Makarcyzk
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
| | - Ilhan Yu
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Eileen N. Li
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
| | - Madalyn R. Fritch
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Qi Gao
- Department of Orthopaedic SurgeryStanford UniversityStanfordCA94305USA
| | - Kek Boon Goh
- Institute of PhysicsUniversity of FreiburgFreiburg79104Germany
- School of EngineeringMonash University MalaysiaSelangor47500Malaysia
| | - Benjamen O'Donnell
- Center for Stem Cell Research and Regenerative MedicineTulane University School of MedicineOrleansLA70112USA
| | - Tingjun Hao
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Peter G. Alexander
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| | - Bhushan Mahadik
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - John P. Fisher
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Stuart B. Goodman
- Department of Orthopaedic SurgeryStanford UniversityStanfordCA94305USA
| | - Bruce A. Bunnell
- Center for Stem Cell Research and Regenerative MedicineTulane University School of MedicineOrleansLA70112USA
- Present address:
Department of Microbiology, Immunology, and GeneticsUniversity of North Texas Health Science CenterFort WorthTX76107USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
- McGowan Institute for Regenerative MedicineUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Present address:
The Chinese University of Hong KongShatinHong Kong SAR999077China
| | - Hang Lin
- Center for Cellular and Molecular EngineeringDepartment of Orthopaedic SurgeryUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
- Department of BioengineeringUniversity of Pittsburgh Swanson School of EngineeringPittsburghPA15260USA
- McGowan Institute for Regenerative MedicineUniversity of Pittsburgh School of MedicinePittsburghPA15219USA
| |
Collapse
|
13
|
Rothbauer M, Reihs EI, Fischer A, Windhager R, Jenner F, Toegel S. A Progress Report and Roadmap for Microphysiological Systems and Organ-On-A-Chip Technologies to Be More Predictive Models in Human (Knee) Osteoarthritis. Front Bioeng Biotechnol 2022; 10:886360. [PMID: 35782494 PMCID: PMC9240813 DOI: 10.3389/fbioe.2022.886360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Osteoarthritis (OA), a chronic debilitating joint disease affecting hundreds of million people globally, is associated with significant pain and socioeconomic costs. Current treatment modalities are palliative and unable to stop the progressive degeneration of articular cartilage in OA. Scientific attention has shifted from the historical view of OA as a wear-and-tear cartilage disorder to its recognition as a whole-joint disease, highlighting the contribution of other knee joint tissues in OA pathogenesis. Despite much progress in the field of microfluidic systems/organs-on-a-chip in other research fields, current in vitro models in use do not yet accurately reflect the complexity of the OA pathophenotype. In this review, we provide: 1) a detailed overview of the most significant recent developments in the field of microsystems approaches for OA modeling, and 2) an OA-pathophysiology-based bioengineering roadmap for the requirements of the next generation of more predictive and authentic microscale systems fit for the purpose of not only disease modeling but also of drug screening to potentially allow OA animal model reduction and replacement in the near future.
Collapse
Affiliation(s)
- Mario Rothbauer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Eva I. Reihs
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Anita Fischer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Florien Jenner
- Veterinary Tissue Engineering and Regenerative Medicine Vienna (VETERM), Equine Surgery Unit, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Toegel
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|
14
|
Gomez-Florit M, Labrador-Rached CJ, Domingues RM, Gomes ME. The tendon microenvironment: Engineered in vitro models to study cellular crosstalk. Adv Drug Deliv Rev 2022; 185:114299. [PMID: 35436570 DOI: 10.1016/j.addr.2022.114299] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
Abstract
Tendinopathy is a multi-faceted pathology characterized by alterations in tendon microstructure, cellularity and collagen composition. Challenged by the possibility of regenerating pathological or ruptured tendons, the healing mechanisms of this tissue have been widely researched over the past decades. However, so far, most of the cellular players and processes influencing tendon repair remain unknown, which emphasizes the need for developing relevant in vitro models enabling to study the complex multicellular crosstalk occurring in tendon microenvironments. In this review, we critically discuss the insights on the interaction between tenocytes and the other tendon resident cells that have been devised through different types of existing in vitro models. Building on the generated knowledge, we stress the need for advanced models able to mimic the hierarchical architecture, cellularity and physiological signaling of tendon niche under dynamic culture conditions, along with the recreation of the integrated gradients of its tissue interfaces. In a forward-looking vision of the field, we discuss how the convergence of multiple bioengineering technologies can be leveraged as potential platforms to develop the next generation of relevant in vitro models that can contribute for a deeper fundamental knowledge to develop more effective treatments.
Collapse
|
15
|
Kahraman E, Ribeiro R, Lamghari M, Neto E. Cutting-Edge Technologies for Inflamed Joints on Chip: How Close Are We? Front Immunol 2022; 13:802440. [PMID: 35359987 PMCID: PMC8960235 DOI: 10.3389/fimmu.2022.802440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling musculoskeletal disorder, with a large impact on the global population, resulting in several limitations on daily activities. In OA, inflammation is frequent and mainly controlled through inflammatory cytokines released by immune cells. These outbalanced inflammatory cytokines cause cartilage extracellular matrix (ECM) degradation and possible growth of neuronal fibers into subchondral bone triggering pain. Even though pain is the major symptom of musculoskeletal diseases, there are still no effective treatments to counteract it and the mechanisms behind these pathologies are not fully understood. Thus, there is an urgent need to establish reliable models for assessing the molecular mechanisms and consequently new therapeutic targets. Models have been established to support this research field by providing reliable tools to replicate the joint tissue in vitro. Studies firstly started with simple 2D culture setups, followed by 3D culture focusing mainly on cell-cell interactions to mimic healthy and inflamed cartilage. Cellular approaches were improved by scaffold-based strategies to enhance cell-matrix interactions as well as contribute to developing mechanically more stable in vitro models. The progression of the cartilage tissue engineering would then profit from the integration of 3D bioprinting technologies as these provide 3D constructs with versatile structural arrangements of the 3D constructs. The upgrade of the available tools with dynamic conditions was then achieved using bioreactors and fluid systems. Finally, the organ-on-a-chip encloses all the state of the art on cartilage tissue engineering by incorporation of different microenvironments, cells and stimuli and pave the way to potentially simulate crucial biological, chemical, and mechanical features of arthritic joint. In this review, we describe the several available tools ranging from simple cartilage pellets to complex organ-on-a-chip platforms, including 3D tissue-engineered constructs and bioprinting tools. Moreover, we provide a fruitful discussion on the possible upgrades to enhance the in vitro systems making them more robust regarding the physiological and pathological modeling of the joint tissue/OA.
Collapse
Affiliation(s)
- Emine Kahraman
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Rua Dr. Roberto Frias, Porto, Portugal
| | - Ricardo Ribeiro
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
16
|
Muenzebrock KA, Kersten V, Alblas J, Garcia JP, Creemers LB. The Added Value of the “Co” in Co-Culture Systems in Research on Osteoarthritis Pathology and Treatment Development. Front Bioeng Biotechnol 2022; 10:843056. [PMID: 35309991 PMCID: PMC8927651 DOI: 10.3389/fbioe.2022.843056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent disease and a major health burden. Its development and progression are influenced by factors such as age, obesity or joint overuse. As a whole organ disease OA affects not only cartilage, bone and synovium but also ligaments, fatty or nervous tissue surrounding the joint. These joint tissues interact with each other and understanding this interaction is important in developing novel treatments. To incorporate and study these interactions in OA research, several co-culture models have evolved. They combine two or more cell types or tissues and investigate the influence of amongst others inflammatory or degenerative stimuli seen in OA. This review focuses on co-cultures and the differential processes occurring in a given tissue or cell as a consequence of being combined with another joint cell type or tissue, and/or the extent to which a co-culture mimics the in vivo processes. Most co-culture models depart from synovial lining and cartilage culture, but also fat pad and bone have been included. Not all of the models appear to reflect the postulated in vivo OA pathophysiology, although some of the discrepancies may indicate current assumptions on this process are not entirely valid. Systematic analysis of the mutual influence the separate compartments in a given model exert on each other and validation against in vivo or ex vivo observation is still largely lacking and would increase their added value as in vitro OA models.
Collapse
|
17
|
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]
|
18
|
Zou Z, Luo X, Chen Z, Zhang YS, Wen C. Emerging microfluidics-enabled platforms for osteoarthritis management: from benchtop to bedside. Theranostics 2022; 12:891-909. [PMID: 34976219 PMCID: PMC8692897 DOI: 10.7150/thno.62685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/17/2021] [Indexed: 11/12/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent debilitating age-related joint degenerative disease. It is a leading cause of pain and functional disability in older adults. Unfortunately, there is no cure for OA once the damage is established. Therefore, it promotes an urgent need for early detection and intervention of OA. Theranostics, combining therapy and diagnosis, emerges as a promising approach for OA management. However, OA theranostics is still in its infancy. Three fundamental needs have to be firstly fulfilled: i) a reliable OA model for disease pathogenesis investigation and drug screening, ii) an effective and precise diagnostic platform, and iii) an advanced fabrication approach for drug delivery and therapy. Meanwhile, microfluidics emerges as a versatile technology to address each of the needs and eventually boost the development of OA theranostics. Therefore, this review focuses on the applications of microfluidics, from benchtop to bedside, for OA modelling and drug screening, early diagnosis, and clinical therapy. We first introduce the basic pathophysiology of OA and point out the major unfilled research gaps in current OA management including lack of disease modelling and drug screening platforms, early diagnostic modalities and disease-modifying drugs and delivery approaches. Accordingly, we then summarize the state-of-the-art microfluidics technology for OA management from in vitro modelling and diagnosis to therapy. Given the existing promising results, we further discuss the future development of microfluidic platforms towards clinical translation at the crossroad of engineering and biomedicine.
Collapse
Affiliation(s)
- Zhou Zou
- Department of Biomedical Engineering, Faculty of Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiaohe Luo
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zhengkun Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
- Currently at Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
19
|
Abstract
AbstractThe multidisciplinary research field of bioprinting combines additive manufacturing, biology and material sciences to create bioconstructs with three-dimensional architectures mimicking natural living tissues. The high interest in the possibility of reproducing biological tissues and organs is further boosted by the ever-increasing need for personalized medicine, thus allowing bioprinting to establish itself in the field of biomedical research, and attracting extensive research efforts from companies, universities, and research institutes alike. In this context, this paper proposes a scientometric analysis and critical review of the current literature and the industrial landscape of bioprinting to provide a clear overview of its fast-changing and complex position. The scientific literature and patenting results for 2000–2020 are reviewed and critically analyzed by retrieving 9314 scientific papers and 309 international patents in order to draw a picture of the scientific and industrial landscape in terms of top research countries, institutions, journals, authors and topics, and identifying the technology hubs worldwide. This review paper thus offers a guide to researchers interested in this field or to those who simply want to understand the emerging trends in additive manufacturing and 3D bioprinting.
Graphic abstract
Collapse
|
20
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Overcoming the Dependence on Animal Models for Osteoarthritis Therapeutics - The Promises and Prospects of In Vitro Models. Adv Healthc Mater 2021; 10:e2100961. [PMID: 34302436 DOI: 10.1002/adhm.202100961] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis (OA) is a musculoskeletal disease characterized by progressive degeneration of osteochondral tissues. Current treatment is restricted to the reduction of pain and loss of function of the joint. To better comprehend the OA pathophysiological conditions, several models are employed, however; there is no consensus on a suitable model. In this review, different in vitro models being developed for possible therapeutic intervention of OA are outlined. Herein, various in vitro OA models starting from 2D model, co-culture model, 3D models, dynamic culture model to advanced technologies-based models such as 3D bioprinting, bioassembly, organoids, and organ-on-chip-based models are discussed with their advantages and disadvantages. Besides, different growth factors, cytokines, and chemicals being utilized for induction of OA condition are reviewed in detail. Furthermore, there is focus on scrutinizing different molecular and possible therapeutic targets for better understanding the mechanisms and OA therapeutics. Finally, the underlying challenges associated with in vitro models are discussed followed by future prospective. Taken together, a comprehensive overview of in vitro OA models, factors to induce OA-like conditions, and intricate molecular targets with the potential to develop personalized osteoarthritis therapeutics in the future with clinical translation is provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Joseph Christakiran Moses
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Nandana Bhardwaj
- Department of Science and Mathematics Indian Institute of Information Technology Guwahati Bongora Guwahati Assam 781015 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
21
|
Tschon M, Contartese D, Pagani S, Borsari V, Fini M. Gender and Sex Are Key Determinants in Osteoarthritis Not Only Confounding Variables. A Systematic Review of Clinical Data. J Clin Med 2021; 10:3178. [PMID: 34300344 PMCID: PMC8303951 DOI: 10.3390/jcm10143178] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/08/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022] Open
Abstract
Many risk factors for osteoarthritis (OA) have been noted, while gender/sex differences have been understated. The work aimed to systematically review literature investigating as primary aim the relationship between gender/sex related discriminants and OA. The search was performed in PubMed, Science Direct and Web of Knowledge in the last 10 years. Inclusion criteria were limited to clinical studies of patients affected by OA in any joints, analyzing as primary aim gender/sex differences. Exclusion criteria were review articles, in vitro, in vivo and ex vivo studies, case series studies and papers in which gender/sex differences were adjusted as confounding variable. Of the 120 records screened, 42 studies were included. Different clinical outcomes were analyzed: morphometric differences, followed by kinematics, pain, functional outcomes after arthroplasty and health care needs of patients. Women appear to use more health care, have higher OA prevalence, clinical pain and inflammation, decreased cartilage volume, physical difficulty, and smaller joint parameters and dimensions, as compared to men. No in-depth studies or mechanistic studies analyzing biomarker differential expressions, molecular pathways and omic profiles were found that might drive preclinical and clinical research towards sex-/gender-oriented protocols.
Collapse
Affiliation(s)
| | - Deyanira Contartese
- Surgical Sciences and Tecnologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (M.T.); (S.P.); (V.B.); (M.F.)
| | | | | | | |
Collapse
|
22
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
23
|
Makarczyk MJ, Gao Q, He Y, Li Z, Gold MS, Hochberg MC, Bunnell BA, Tuan RS, Goodman SB, Lin H. Current Models for Development of Disease-Modifying Osteoarthritis Drugs. Tissue Eng Part C Methods 2021; 27:124-138. [PMID: 33403944 PMCID: PMC8098772 DOI: 10.1089/ten.tec.2020.0309] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling disease that affects millions of people worldwide. Symptom-alleviating treatments exist, although none with long-term efficacy. Furthermore, there are currently no disease-modifying OA drugs (DMOADs) with demonstrated efficacy in OA patients, which is, in part, attributed to a lack of full understanding of the pathogenesis of OA. The inability to translate findings from basic research to clinical applications also highlights the deficiencies in the available OA models at simulating the clinically relevant pathologies and responses to treatments in humans. In this review, the current status in the development of DMOADs will be first presented, with special attention to those in Phase II-IV clinical trials. Next, current in vitro, ex vivo, and in vivo OA models are summarized and the respective advantages and disadvantages of each are highlighted. Of note, the development and application of microphysiological or tissue-on-a-chip systems for modeling OA in humans are presented and the issues that need to be addressed in the future are discussed. Microphysiological systems should be given serious consideration for their inclusion in the DMOAD development pipeline, both for their ability to predict drug safety and efficacy in human clinical trials at present, as well as for their potential to serve as a test platform for personalized medicine. Impact statement At present, no disease-modifying osteoarthritis (OA) drugs (DMOADs) have been approved for widespread clinical use by regulatory bodies. The failure of developing effective DMOADs is likely owing to multiple factors, not the least of which are the intrinsic differences between the intact human knee joint and the preclinical models. This work summarizes the current OA models for the development of DMOADs, discusses the advantages/disadvantages of each, and then proposes future model development to aid in the discovery of effective and personalized DMOADs. The review also highlights the microphysiological systems, which are emerging as a new platform for drug development.
Collapse
Affiliation(s)
- Meagan J. Makarczyk
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, California, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael S. Gold
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark C. Hochberg
- Department of Medicine and Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, California, USA
- Department of Bioengineering, Stanford University, California, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Salgado C, Jordan O, Allémann E. Osteoarthritis In Vitro Models: Applications and Implications in Development of Intra-Articular Drug Delivery Systems. Pharmaceutics 2021; 13:60. [PMID: 33466397 PMCID: PMC7824837 DOI: 10.3390/pharmaceutics13010060] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a complex multi-target disease with an unmet medical need for the development of therapies that slow and potentially revert disease progression. Intra-articular (IA) delivery has seen a surge in osteoarthritis research in recent years. As local administration of molecules, this represents a way to circumvent systemic drug delivery struggles. When developing intra-articular formulations, the main goals are a sustained and controlled release of therapeutic drug doses, taking into account carrier choice, drug molecule, and articular joint tissue target. Therefore, the selection of models is critical when developing local administration formulation in terms of accurate outcome assessment, target and off-target effects and relevant translation to in vivo. The current review highlights the applications of OA in vitro models in the development of IA formulation by means of exploring their advantages and disadvantages. In vitro models are essential in studies of OA molecular pathways, understanding drug and target interactions, assessing cytotoxicity of carriers and drug molecules, and predicting in vivo behaviors. However, further understanding of molecular and tissue-specific intricacies of cellular models for 2D and 3D needs improvement to accurately portray in vivo conditions.
Collapse
Affiliation(s)
- Carlota Salgado
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
25
|
A Human Osteochondral Tissue Model Mimicking Cytokine-Induced Key Features of Arthritis In Vitro. Int J Mol Sci 2020; 22:ijms22010128. [PMID: 33374446 PMCID: PMC7794893 DOI: 10.3390/ijms22010128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/15/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
Adequate tissue engineered models are required to further understand the (patho)physiological mechanism involved in the destructive processes of cartilage and subchondral bone during rheumatoid arthritis (RA). Therefore, we developed a human in vitro 3D osteochondral tissue model (OTM), mimicking cytokine-induced cellular and matrix-related changes leading to cartilage degradation and bone destruction in order to ultimately provide a preclinical drug screening tool. To this end, the OTM was engineered by co-cultivation of mesenchymal stromal cell (MSC)-derived bone and cartilage components in a 3D environment. It was comprehensively characterized on cell, protein, and mRNA level. Stimulating the OTM with pro-inflammatory cytokines, relevant in RA (tumor necrosis factor α, interleukin-6, macrophage migration inhibitory factor), caused cell- and matrix-related changes, resulting in a significantly induced gene expression of lactate dehydrogenase A, interleukin-8 and tumor necrosis factor α in both, cartilage and bone, while the matrix metalloproteases 1 and 3 were only induced in cartilage. Finally, application of target-specific drugs prevented the induction of inflammation and matrix-degradation. Thus, we here provide evidence that our human in vitro 3D OTM mimics cytokine-induced cell- and matrix-related changes—key features of RA—and may serve as a preclinical tool for the evaluation of both new targets and potential drugs in a more translational setup.
Collapse
|
26
|
Rothbauer M, Höll G, Eilenberger C, Kratz SRA, Farooq B, Schuller P, Olmos Calvo I, Byrne RA, Meyer B, Niederreiter B, Küpcü S, Sevelda F, Holinka J, Hayden O, Tedde SF, Kiener HP, Ertl P. Monitoring tissue-level remodelling during inflammatory arthritis using a three-dimensional synovium-on-a-chip with non-invasive light scattering biosensing. LAB ON A CHIP 2020; 20:1461-1471. [PMID: 32219235 DOI: 10.1039/c9lc01097a] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Rheumatoid arthritis is a chronic, systemic joint disease in which an autoimmune response translates into an inflammatory attack resulting in joint damage, disability and decreased quality of life. Despite recent introduction of therapeutic agents such as anti-TNFα, even the best current therapies fail to achieve disease remission in most arthritis patients. Therefore, research into the mechanisms governing the destructive inflammatory process in rheumatoid arthritis is of great importance and may reveal novel strategies for the therapeutic interventions. To gain deeper insight into its pathogensis, we have developed for the first time a three-dimensional synovium-on-a-chip system in order to monitor the onset and progression of inflammatory synovial tissue responses. In our study, patient-derived primary synovial organoids are cultivated on a single chip platform containing embedded organic-photodetector arrays for over a week in the absence and presence of tumor-necrosis-factor. Using a label-free and non-invasive optical light-scatter biosensing strategy inflammation-induced 3D tissue-level architectural changes were already detected after two days. We demonstrate that the integration of complex human synovial organ cultures in a lab-on-a-chip provides reproducible and reliable information on how systemic stress factors affect synovial tissue architectures.
Collapse
Affiliation(s)
- Mario Rothbauer
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Piluso S, Labet M, Zhou C, Seo JW, Thielemans W, Patterson J. Engineered Three-Dimensional Microenvironments with Starch Nanocrystals as Cell-Instructive Materials. Biomacromolecules 2019; 20:3819-3830. [PMID: 31490664 DOI: 10.1021/acs.biomac.9b00907] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Naturally, cells reside in three-dimensional (3D) microenvironments composed of biopolymers that guide cellular behavior via topographical features as well as through mechanical and biochemical cues. However, most studies describing the influence of topography on cells' behavior are performed on rigid and synthetic two-dimensional substrates. To design systems that more closely resemble native microenvironments, herein we develop 3D nanocomposite hydrogels consisting of starch nanocrystals (SNCs) embedded in a gelatin matrix. The incorporation of different concentrations of SNCs (0.05, 0.2, and 0.5 wt %) results in an increase of compressive modulus when compared to hydrogels without SNCs, without affecting the swelling ratio, thus providing a tunable system. Confirming the cytocompatibility of the novel composites, the viability of encapsulated L929 fibroblasts is >90% in all hydrogels. The cellular metabolic activity and DNA content are similar for all formulations and increase over time, indicating that the fibroblasts proliferate within the hydrogels. After 4 d of culture, Live/Dead staining and F-actin/nuclei staining show that the encapsulated fibroblasts develop an elongated morphology in the hydrogels. On the other hand, encapsulated chondrogenic progenitor ATDC5 cells also maintain a viability around 90% but display a round morphology, especially in the hydrogels with SNCs, indicating a potential application of the materials for cartilage tissue engineering. We believe that topographical and mechanical cues within 3D microenvironments can be a powerful tool to instruct cells' behavior and that the developed gelatin/SNC nanocomposite warrants further study.
Collapse
Affiliation(s)
- Susanna Piluso
- Department of Materials Engineering , KU Leuven , 3001 Leuven , Belgium
| | - Marianne Labet
- Renewable Materials and Nanotechnology Research Group, Department of Chemical Engineering , KU Leuven , Campus Kulak Kortrijk , 8500 Kortrijk , Belgium
| | - Chen Zhou
- Department of Materials Engineering , KU Leuven , 3001 Leuven , Belgium
| | - Jin Won Seo
- Department of Materials Engineering , KU Leuven , 3001 Leuven , Belgium
| | - Wim Thielemans
- Renewable Materials and Nanotechnology Research Group, Department of Chemical Engineering , KU Leuven , Campus Kulak Kortrijk , 8500 Kortrijk , Belgium
| | | |
Collapse
|