1
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024:e2401307. [PMID: 39175382 DOI: 10.1002/adhm.202401307] [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: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| | - Janis Locs
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| |
Collapse
|
2
|
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
|
3
|
Quek J, Vizetto-Duarte C, Teoh SH, Choo Y. Towards Stem Cell Therapy for Critical-Sized Segmental Bone Defects: Current Trends and Challenges on the Path to Clinical Translation. J Funct Biomater 2024; 15:145. [PMID: 38921519 PMCID: PMC11205181 DOI: 10.3390/jfb15060145] [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: 04/24/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
The management and reconstruction of critical-sized segmental bone defects remain a major clinical challenge for orthopaedic clinicians and surgeons. In particular, regenerative medicine approaches that involve incorporating stem cells within tissue engineering scaffolds have great promise for fracture management. This narrative review focuses on the primary components of bone tissue engineering-stem cells, scaffolds, the microenvironment, and vascularisation-addressing current advances and translational and regulatory challenges in the current landscape of stem cell therapy for critical-sized bone defects. To comprehensively explore this research area and offer insights for future treatment options in orthopaedic surgery, we have examined the latest developments and advancements in bone tissue engineering, focusing on those of clinical relevance in recent years. Finally, we present a forward-looking perspective on using stem cells in bone tissue engineering for critical-sized segmental bone defects.
Collapse
Affiliation(s)
- Jolene Quek
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Catarina Vizetto-Duarte
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Swee Hin Teoh
- Centre for Advanced Medical Engineering, College of Materials Science and Engineering, Hunan University, Changsha 410012, China
| | - Yen Choo
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| |
Collapse
|
4
|
Lai Z, Niu X, Chen X, Lu F, Zhang Y, Yuan Y. Composite Microparticles of Fat Graft and GFR Matrigel Improved Volume Retention by Promoting Cell Migration and Vessel Regeneration. Aesthetic Plast Surg 2024; 48:1993-2001. [PMID: 38302709 DOI: 10.1007/s00266-022-03145-6] [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: 07/16/2022] [Accepted: 10/09/2022] [Indexed: 02/03/2024]
Abstract
BACKGROUND The retention volume of autologous fat grafts decreases after transplantation due to limited nutrition infiltration and insufficient blood supply. Structural fat grafts and the 3M (multipoint, multitunnel, and multilayer) injection technique have been considered to improve the survival of grafts; however, it is difficult for surgeons to practice in the clinic because grafts tend to gather into a cluster, especially in large volume fat grafting. Therefore, we hypothesize that prefabricated microparticle fat grafts (PFMG) may improve the retention rate. METHODS The C57BL/6 mouse fat particles were embedded in growth factor-reduced (GFR)-Matrigel to detect cell migration by immunofluorescence staining in vitro. PFMG was prepared by mixing mouse fat particles and GFR Matrigel in a 1:1 volume ratio and injected subcutaneously into C57BL/6 mice. Fat particles mixed with PBS in equal volume served as control group. The grafts were harvested at 1, 4, 8, and 12 weeks after sacrifice. The retention rate of grafts at each time point was measured, and the structural alterations were detected by SEM. Fat necrosis and blood vessel density were evaluated by histological analysis. RESULTS CD34+ cells are migrated from the PFMG and formed a tree-like tubular network in the in vitro study. The retention rate was higher in the PFMG group than in the control group at week 12 (38% vs. 30%, p < 0.05). After transplantation, the dissociated structure of fat particles was maintained in PFMG by SEM analysis. Histological analysis of PFMG confirmed less fat necrosis and more blood vessel density in the PFMG group at the early stage than in the control group. The GFR Matrigel was displaced by adipose tissue with time. CONCLUSIONS In this study, we developed a novel fat grafting method, PFMG that dispersed fat grafts and maintained the structure after transplantation. High volume retention volume of PFMG was achieved by promoting cell migration and vessel regeneration. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Zhuhao Lai
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, 219 Moganshan Road, 310005, Hangzhou, People's Republic of China
| | - Xingtang Niu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xihang Chen
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yuchen Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yi Yuan
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
5
|
Toni R, Barbaro F, Di Conza G, Zini N, Remaggi G, Elviri L, Spaletta G, Quarantini E, Quarantini M, Mosca S, Caravelli S, Mosca M, Ravanetti F, Sprio S, Tampieri A. A bioartificial and vasculomorphic bone matrix-based organoid mimicking microanatomy of flat and short bones. J Biomed Mater Res B Appl Biomater 2024; 112:e35329. [PMID: 37898921 DOI: 10.1002/jbm.b.35329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 10/31/2023]
Abstract
We engineered an in vitro model of bioartificial 3D bone organoid consistent with an anatomical and vascular microenvironment common to mammalian flat and short bones. To achieve this, we chose the decellularized-decalcified matrix of the adult male rat scapula, implemented with the reconstruction of its intrinsic vessels, obtained through an original intravascular perfusion with polylevolactic (PLLA), followed by coating of the PLLA-fabricated vascularization with rat tail collagen. As a result, the 3D bone and vascular geometry of the native bone cortical and cancellous compartments was reproduced, and the rat tail collagen-PLLA biomaterial could in vitro act as a surrogate of the perivascular extracellular matrix (ECM) around the wall of the biomaterial-reconstituted cancellous vessels. As a proof-of-concept of cell compatibility and site-dependent osteoinductive properties of this bioartificial 3D construct, we show that it in vitro leads to a time-dependent microtopographic positioning of rat mesenchymal stromal cells (MSCs), initiating an osteogenic fate in relation to the bone compartment. In addition, coating of PLLA-reconstructed vessels with rat tail collagen favored perivascular attachment and survival of MSC-like cells (mouse embryonic fibroblasts), confirming its potentiality as a perivascular stroma for triggering competence of seeded MSCs. Finally, in vivo radiographic topography of bone lesions in the human jaw and foot tarsus of subjects with primary osteoporosis revealed selective bone cortical versus cancellous involvement, suggesting usefulness of a human 3D bone organoid engineered with the same principles of our rat organoid, to in vitro investigate compartment-dependent activities of human MSC in flat and short bones under experimental osteoporotic challenge. We conclude that our 3D bioartificial construct offers a reliable replica of flat and short bones microanatomy, and promises to help in building a compartment-dependent mechanistic perspective of bone remodeling, including the microtopographic dysregulation of osteoporosis.
Collapse
Affiliation(s)
- Roberto Toni
- ISSMC, CNR, Faenza, Italy
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center-Tufts University School of Medicine, Boston, Massachusetts, USA
- Academy of Sciences of the Institute of Bologna, Section IV-Medical Sciences, Bologna, Italy
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic-OSTEONET (Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies) and Odontostomatology Units, Galliera Medical Center, San Venanzio di Galliera (BO), Italy
| | - Fulvio Barbaro
- Department of Medicine and Surgery-DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine-BIOMED, University of Parma, Parma, Italy
| | - Giusy Di Conza
- Department of Medicine and Surgery-DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine-BIOMED, University of Parma, Parma, Italy
| | - Nicoletta Zini
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giulia Remaggi
- Food and Drug Department, University of Parma, Parma, Italy
| | - Lisa Elviri
- Food and Drug Department, University of Parma, Parma, Italy
| | - Giulia Spaletta
- Department of Statistical Sciences, University of Bologna, Bologna, Italy
| | - Enrico Quarantini
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic-OSTEONET (Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies) and Odontostomatology Units, Galliera Medical Center, San Venanzio di Galliera (BO), Italy
| | - Marco Quarantini
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic-OSTEONET (Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies) and Odontostomatology Units, Galliera Medical Center, San Venanzio di Galliera (BO), Italy
| | - Salvatore Mosca
- Course on Disorders of the Locomotor System, Fellow Program in Orthopaedics and Traumatology, University Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Caravelli
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Massimiliano Mosca
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Ravanetti
- Department of Veterinary Medical Sciences, Section of Anatomy, University of Parma, Parma, Italy
| | | | | |
Collapse
|
6
|
Shan C, Xia Y, Wu Z, Zhao J. HIF-1α and periodontitis: Novel insights linking host-environment interplay to periodontal phenotypes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 184:50-78. [PMID: 37769974 DOI: 10.1016/j.pbiomolbio.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Periodontitis, the sixth most prevalent epidemic disease globally, profoundly impacts oral aesthetics and masticatory functionality. Hypoxia-inducible factor-1α (HIF-1α), an oxygen-dependent transcriptional activator, has emerged as a pivotal regulator in periodontal tissue and alveolar bone metabolism, exerts critical functions in angiogenesis, erythropoiesis, energy metabolism, and cell fate determination. Numerous essential phenotypes regulated by HIF are intricately associated with bone metabolism in periodontal tissues. Extensive investigations have highlighted the central role of HIF and its downstream target genes and pathways in the coupling of angiogenesis and osteogenesis. Within this concise perspective, we comprehensively review the cellular phenotypic alterations and microenvironmental dynamics linking HIF to periodontitis. We analyze current research on the HIF pathway, elucidating its impact on bone repair and regeneration, while unraveling the involved cellular and molecular mechanisms. Furthermore, we briefly discuss the potential application of targeted interventions aimed at HIF in the field of bone tissue regeneration engineering. This review expands our biological understanding of the intricate relationship between the HIF gene and bone angiogenesis in periodontitis and offers valuable insights for the development of innovative therapies to expedite bone repair and regeneration.
Collapse
Affiliation(s)
- Chao Shan
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - YuNing Xia
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Zeyu Wu
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Jin Zhao
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China; Xinjiang Uygur Autonomous Region Institute of Stomatology, Ürümqi, China.
| |
Collapse
|
7
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
8
|
Stein M, Elefteriou F, Busse B, Fiedler IA, Kwon RY, Farell E, Ahmad M, Ignatius A, Grover L, Geris L, Tuckermann J. Why Animal Experiments Are Still Indispensable in Bone Research: A Statement by the European Calcified Tissue Society. J Bone Miner Res 2023; 38:1045-1061. [PMID: 37314012 PMCID: PMC10962000 DOI: 10.1002/jbmr.4868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 05/03/2023] [Accepted: 06/09/2023] [Indexed: 06/15/2023]
Abstract
Major achievements in bone research have always relied on animal models and in vitro systems derived from patient and animal material. However, the use of animals in research has drawn intense ethical debate and the complete abolition of animal experimentation is demanded by fractions of the population. This phenomenon is enhanced by the reproducibility crisis in science and the advance of in vitro and in silico techniques. 3D culture, organ-on-a-chip, and computer models have improved enormously over the last few years. Nevertheless, the overall complexity of bone tissue cross-talk and the systemic and local regulation of bone physiology can often only be addressed in entire vertebrates. Powerful genetic methods such as conditional mutagenesis, lineage tracing, and modeling of the diseases enhanced the understanding of the entire skeletal system. In this review endorsed by the European Calcified Tissue Society (ECTS), a working group of investigators from Europe and the US provides an overview of the strengths and limitations of experimental animal models, including rodents, fish, and large animals, as well the potential and shortcomings of in vitro and in silico technologies in skeletal research. We propose that the proper combination of the right animal model for a specific hypothesis and state-of-the-art in vitro and/or in silico technology is essential to solving remaining important questions in bone research. This is crucial for executing most efficiently the 3R principles to reduce, refine, and replace animal experimentation, for enhancing our knowledge of skeletal biology, and for the treatment of bone diseases that affect a large part of society. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Merle Stein
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Florent Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), University Medical Center Hamburg-Eppendorf, Germany
| | - Imke A.K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), University Medical Center Hamburg-Eppendorf, Germany
| | - Ronald Young Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, USA and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Eric Farell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Mubashir Ahmad
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Liam Grover
- Healthcare Technologies Institute, Institute of Translational MedicineHeritage Building Edgbaston, Birmingham
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In Silico Medicine, University of Liège, Liège, Belgium
- Skeletal Biology & Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| |
Collapse
|
9
|
Micalet A, Pape J, Bakkalci D, Javanmardi Y, Hall C, Cheema U, Moeendarbary E. Evaluating the Impact of a Biomimetic Mechanical Environment on Cancer Invasion and Matrix Remodeling. Adv Healthc Mater 2023; 12:e2201749. [PMID: 36333907 PMCID: PMC11468596 DOI: 10.1002/adhm.202201749] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/10/2022] [Indexed: 10/13/2024]
Abstract
The stiffness of tumors and their host tissues is much higher than most hydrogels, which are conventionally used to study in vitro cancer progression. The tumoroid assay is an engineered 3D in vitro tumor model that allows investigation of cancer cell invasion in an environment that is biomimetic in terms of extracellular matrix (ECM) composition and stiffness. Using this model, the change in matrix stiffness by epithelial colorectal cancer cells is systematically characterized by atomic force microscopy indentation tests. Less invasive epithelial cancer cells stiffen the tumor microenvironment while highly aggressive epithelial cancer cells show significant softening of the tumor microenvironment. Changes in stiffness are attributed to both cell-generated active forces as well as ECM degradation and remodeling. The degradation is in part attributed to the enzymatic activity of matrix metalloproteinases (MMPs) as demonstrated by the significant expression of MMP-2 and MMP-9 at both gene and protein levels. Targeting MMP activity through broad-spectrum drug inhibition (BB-94) reverses the changes in stiffness and also decreases cancer cell invasion. These results promote the idea of using mechano-based cancer therapies such as MMP inhibition.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Judith Pape
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Deniz Bakkalci
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Yousef Javanmardi
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Chloe Hall
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Umber Cheema
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Emad Moeendarbary
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| |
Collapse
|
10
|
Javanmardi Y, Agrawal A, Malandrino A, Lasli S, Chen M, Shahreza S, Serwinski B, Cammoun L, Li R, Jorfi M, Djordjevic B, Szita N, Spill F, Bertazzo S, Sheridan GK, Shenoy V, Calvo F, Kamm R, Moeendarbary E. Endothelium and Subendothelial Matrix Mechanics Modulate Cancer Cell Transendothelial Migration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206554. [PMID: 37051804 PMCID: PMC10238207 DOI: 10.1002/advs.202206554] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/19/2023] [Indexed: 06/04/2023]
Abstract
Cancer cell extravasation, a key step in the metastatic cascade, involves cancer cell arrest on the endothelium, transendothelial migration (TEM), followed by the invasion into the subendothelial extracellular matrix (ECM) of distant tissues. While cancer research has mostly focused on the biomechanical interactions between tumor cells (TCs) and ECM, particularly at the primary tumor site, very little is known about the mechanical properties of endothelial cells and the subendothelial ECM and how they contribute to the extravasation process. Here, an integrated experimental and theoretical framework is developed to investigate the mechanical crosstalk between TCs, endothelium and subendothelial ECM during in vitro cancer cell extravasation. It is found that cancer cell actin-rich protrusions generate complex push-pull forces to initiate and drive TEM, while transmigration success also relies on the forces generated by the endothelium. Consequently, mechanical properties of the subendothelial ECM and endothelial actomyosin contractility that mediate the endothelial forces also impact the endothelium's resistance to cancer cell transmigration. These results indicate that mechanical features of distant tissues, including force interactions between the endothelium and the subendothelial ECM, are key determinants of metastatic organotropism.
Collapse
Affiliation(s)
- Yousef Javanmardi
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
| | - Ayushi Agrawal
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
| | - Andrea Malandrino
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Biomaterials, Biomechanics and Tissue Engineering GroupDepartment of Materials Science and Engineering and Research Center for Biomedical EngineeringUniversitat Politécnica de Catalunya (UPC)08019BarcelonaSpain
| | - Soufian Lasli
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
| | - Michelle Chen
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Somayeh Shahreza
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
| | - Bianca Serwinski
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
- 199 Biotechnologies LtdGloucester RoadLondonW2 6LDUK
| | - Leila Cammoun
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ran Li
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mehdi Jorfi
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Boris Djordjevic
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
- 199 Biotechnologies LtdGloucester RoadLondonW2 6LDUK
| | - Nicolas Szita
- Department of Biochemical EngineeringUniversity College LondonLondonWC1E 6BTUK
| | - Fabian Spill
- School of MathematicsUniversity of BirminghamEdgbastonBirminghamB152TSUK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonWC1E 6BTUK
| | - Graham K Sheridan
- School of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamNG7 2UHUK
| | - Vivek Shenoy
- Department of Materials Science and EngineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria)Santander39011Spain
| | - Roger Kamm
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Emad Moeendarbary
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
11
|
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: 2.0] [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
|
12
|
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: 11] [Impact Index Per Article: 11.0] [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
|
13
|
Watson SA, Javanmardi Y, Zanieri L, Shahreza S, Ragazzini R, Bonfanti P, Moeendarbary E. Integrated role of human thymic stromal cells in hematopoietic stem cell extravasation. Bioeng Transl Med 2023; 8:e10454. [PMID: 36925684 PMCID: PMC10013751 DOI: 10.1002/btm2.10454] [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: 06/10/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
The human thymus is the site of T-cell maturation and induction of central tolerance. Hematopoietic stem cell (HSC)-derived progenitors are recruited to the thymus from the fetal liver during early prenatal development and from bone marrow at later stages and postnatal life. The mechanism by which HSCs are recruited to the thymus is poorly understood in humans, though mouse models have indicated the critical role of thymic stromal cells (TSC). Here, we developed a 3D microfluidic assay based on human cells to model HSC extravasation across the endothelium into the extracellular matrix. We found that the presence of human TSC consisting of cultured thymic epithelial cells (TEC) and interstitial cells (TIC) increases the HSC extravasation rates by 3-fold. Strikingly, incorporating TEC or TIC alone is insufficient to perturb HSC extravasation rates. Furthermore, we identified complex gene expressions from interactions between endothelial cells, TEC and TIC modulates the HSCs extravasation. Our results suggest that comprehensive signaling from the complex thymic microenvironment is crucial for thymus seeding and that our system will allow manipulation of these signals with the potential to increase thymocyte migration in a therapeutic setting.
Collapse
Affiliation(s)
- Sara A. Watson
- Department of Mechanical EngineeringUCLLondonUK
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
| | | | - Luca Zanieri
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | | - Roberta Ragazzini
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | - Paola Bonfanti
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | |
Collapse
|
14
|
Soto Veliz D, Lin K, Sahlgren C. Organ-on-a-chip technologies for biomedical research and drug development: A focus on the vasculature. SMART MEDICINE 2023; 2:e20220030. [PMID: 37089706 PMCID: PMC7614466 DOI: 10.1002/smmd.20220030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/20/2023] [Indexed: 04/25/2023]
Abstract
Current biomedical models fail to replicate the complexity of human biology. Consequently, almost 90% of drug candidates fail during clinical trials after decades of research and billions of investments in drug development. Despite their physiological similarities, animal models often misrepresent human responses, and instead, trigger ethical and societal debates regarding their use. The overall aim across regulatory entities worldwide is to replace, reduce, and refine the use of animal experimentation, a concept known as the Three Rs principle. In response, researchers develop experimental alternatives to improve the biological relevance of in vitro models through interdisciplinary approaches. This article highlights the emerging organ-on-a-chip technologies, also known as microphysiological systems, with a focus on models of the vasculature. The cardiovascular system transports all necessary substances, including drugs, throughout the body while in charge of thermal regulation and communication between other organ systems. In addition, we discuss the benefits, limitations, and challenges in the widespread use of new biomedical models. Coupled with patient-derived induced pluripotent stem cells, organ-on-a-chip technologies are the future of drug discovery, development, and personalized medicine.
Collapse
Affiliation(s)
- Diosangeles Soto Veliz
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Kai‐Lan Lin
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
| | - Cecilia Sahlgren
- Faculty of Science and EngineeringCell Biology, Åbo Akademi UniversityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CenterÅbo Akademi University and University of TurkuTurkuFinland
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoventhe Netherlands
| |
Collapse
|
15
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
16
|
Associated changes in stiffness of collagen scaffolds during osteoblast mineralisation and bone formation. BMC Res Notes 2022; 15:310. [PMID: 36153566 PMCID: PMC9509582 DOI: 10.1186/s13104-022-06203-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Engineering bone in 3D is important for both regenerative medicine purposes and for the development of accurate in vitro models of bone tissue. The changing material stiffness of bone tissue had not yet been monitored throughout the process of mineralisation and bone nodule formation by osteoblasts either during in vitro engineering or in development perspective. Results Within this short research note, stiffness changes (Young’s modulus) during in vitro bone formation by primary osteoblasts in dense collagen scaffolds were monitored using atomic force microscopy. Data analysis revealed significant stiffening of 3D bone cultures at day 5 and 8 that was correlated with the onset of mineral deposition (p < 0.00005).
Collapse
|
17
|
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: 9] [Impact Index Per Article: 4.5] [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
|
18
|
Esteki MH, Malandrino A, Alemrajabi AA, Sheridan GK, Charras G, Moeendarbary E. Poroelastic osmoregulation of living cell volume. iScience 2021; 24:103482. [PMID: 34927026 PMCID: PMC8649806 DOI: 10.1016/j.isci.2021.103482] [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: 01/28/2021] [Revised: 08/19/2021] [Accepted: 11/19/2021] [Indexed: 11/22/2022] Open
Abstract
Cells maintain their volume through fine intracellular osmolarity regulation. Osmotic challenges drive fluid into or out of cells causing swelling or shrinkage, respectively. The dynamics of cell volume changes depending on the rheology of the cellular constituents and on how fast the fluid permeates through the membrane and cytoplasm. We investigated whether and how poroelasticity can describe volume dynamics in response to osmotic shocks. We exposed cells to osmotic perturbations and used defocusing epifluorescence microscopy on membrane-attached fluorescent nanospheres to track volume dynamics with high spatiotemporal resolution. We found that a poroelastic model that considers both geometrical and pressurization rates captures fluid-cytoskeleton interactions, which are rate-limiting factors in controlling volume changes at short timescales. Linking cellular responses to osmotic shocks and cell mechanics through poroelasticity can predict the cell state in health, disease, or in response to novel therapeutics. Cell height changes can be finely captured by defocusing microscopy Water permeation and cellular deformability regulate dynamics of cell volume changes Poroelasticity describes the dynamics of cell volume changes The response of cell to hypo or hyperosmotic shocks are modeled by poroelasticity
Collapse
Affiliation(s)
- Mohammad Hadi Esteki
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan, Iran.,Department of Mechanical Engineering, University College London, London, UK
| | - Andrea Malandrino
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Ali Akbar Alemrajabi
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Graham K Sheridan
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London, UK.,Department of Cell and Developmental Biology, University College London, London, UK.,Institute for the Physics of Living Systems, University College London, London, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
19
|
Javanmardi Y, Colin-York H, Szita N, Fritzsche M, Moeendarbary E. Quantifying cell-generated forces: Poisson's ratio matters. COMMUNICATIONS PHYSICS 2021; 4:237. [PMID: 34841089 PMCID: PMC7612038 DOI: 10.1038/s42005-021-00740-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 10/14/2021] [Indexed: 05/09/2023]
Abstract
Quantifying mechanical forces generated by cellular systems has led to key insights into a broad range of biological phenomena from cell adhesion to immune cell activation. Traction force microscopy (TFM), the most widely employed force measurement methodology, fundamentally relies on knowledge of the force-displacement relationship and mechanical properties of the substrate. Together with the elastic modulus, the Poisson's ratio is a basic material property that to date has largely been overlooked in TFM. Here, we evaluate the sensitivity of TFM to Poisson's ratio by employing a series of computer simulations and experimental data analysis. We demonstrate how applying the correct Poisson's ratio is important for accurate force reconstruction and develop a framework for the determination of error levels resulting from the misestimation of the Poisson's ratio. In addition, we provide experimental estimation of the Poisson's ratios of elastic substrates commonly applied in TFM. Our work thus highlights the role of Poisson's ratio underpinning cellular force quantification studied across many biological systems.
Collapse
Affiliation(s)
- Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Huw Colin-York
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Nicolas Szita
- Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Marco Fritzsche
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK
- Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
20
|
Laschke MW, Menger MD. The simpler, the better: tissue vascularization using the body's own resources. Trends Biotechnol 2021; 40:281-290. [PMID: 34404555 DOI: 10.1016/j.tibtech.2021.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
Tissue regeneration is crucially dependent on sufficient vascularization. In regenerative medicine, this can be effectively achieved by autologous vascularization strategies using the body's own resources. These strategies include the administration of blood-derived factor preparations, adipose tissue-based vascularization, and the in situ engineering of vascularized tissue. Due to their simplicity, the translation of these strategies into clinical practice is easier in terms of feasibility, safety requirements, and regulatory hurdles compared with complex and time-consuming procedures involving intensive cell manipulation. Hence, they are close to clinical application or are already being used to successfully treat patients by distinct personalized medicine concepts.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany.
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| |
Collapse
|