101
|
Douillet C, Nicodeme M, Hermant L, Bergeron V, Guillemot F, Fricain JC, Oliveira H, Garcia M. From local to global matrix organization by fibroblasts: a 4D laser-assisted bioprinting approach. Biofabrication 2021; 14. [PMID: 34875632 DOI: 10.1088/1758-5090/ac40ed] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/07/2021] [Indexed: 11/11/2022]
Abstract
Fibroblasts and myofibroblasts play a central role in skin homeostasis through dermal organization and maintenance. Nonetheless, the dynamic interactions between (myo)fibroblasts and the extracellular matrix (ECM) remain poorly exploited in skin repair strategies. Indeed, there is still an unmet need for soft tissue models allowing to study the spatial-temporal remodeling properties of (myo)fibroblasts. In vivo, wound healing studies in animals are limited by species specificity. In vitro, most models rely on collagen gels reorganized by randomly distributed fibroblasts. But biofabrication technologies have significantly evolved over the past ten years. High-resolution bioprinting now allows to investigate various cellular micropatterns and the emergent tissue organizations over time. In order to harness the full dynamic properties of cells and active biomaterials, it is essential to consider "time" as the 4th dimension in soft tissue design. Following this 4D bioprinting approach, we aimed to develop a novel model that could replicate fibroblast dynamic remodeling in vitro. For this purpose, (myo)fibroblasts were patterned on collagen gels with laser-assisted bioprinting (LAB) to study the generated matrix deformations and reorganizations. First, distinct populations, mainly composed of fibroblasts or myofibroblasts, were established in vitro to account for the variety of fibroblastic remodeling properties. Then, LAB was used to organize both populations on collagen gels in even isotropic patterns with high resolution, high density and high viability. With maturation, bioprinted patterns of fibroblasts and myofibroblasts reorganized into dispersed or aggregated cells, respectively. Stress-release contraction assays revealed that these phenotype-specific pattern maturations were associated with distinct lattice tension states. The two populations were then patterned in anisotropic rows in order to direct the cell-generated deformations and to orient global matrix remodeling. Only maturation of anisotropic fibroblast patterns, but not myofibroblasts, resulted in collagen anisotropic reorganizations both at tissue-scale, with lattice contraction, and at microscale, with embedded microbead displacements. Following a 4D bioprinting approach, LAB patterning enabled to elicit and orient the dynamic matrix remodeling mechanisms of distinct fibroblastic populations and organizations on collagen. For future studies, this method provides a new versatile tool to investigate in vitro dermal organizations and properties, processes of remodeling in healing, and new treatment opportunities.
Collapse
Affiliation(s)
- Camille Douillet
- Bioingénierie tissulaire, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, Aquitaine, 33076, FRANCE
| | - Marc Nicodeme
- Poietis, 27 Allée Charles Darwin, Pessac, 33600, FRANCE
| | - Loïc Hermant
- Poietis, 27 Allée Charles Darwin, Pessac, 33600, FRANCE
| | | | | | - Jean-Christophe Fricain
- Bioingénierie tissulaire, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, 33076, FRANCE
| | - Hugo Oliveira
- Bioingénierie tissulaire, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, 33076, FRANCE
| | - Mikael Garcia
- Poietis, 27 Allée Charles Darwin, Pessac, 33600, FRANCE
| |
Collapse
|
102
|
Cummins KA, Bitterman PB, Tschumperlin DJ, Wood DK. A scalable 3D tissue culture pipeline to enable functional therapeutic screening for pulmonary fibrosis. APL Bioeng 2021; 5:046102. [PMID: 34805716 PMCID: PMC8598262 DOI: 10.1063/5.0054967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease targeting the alveolar gas exchange apparatus, leading to death by asphyxiation. IPF progresses on a tissue scale through aberrant matrix remodeling, enhanced cell contraction, and subsequent microenvironment densification. Although two pharmaceuticals modestly slow progression, IPF patient survival averages less than 5 years. A major impediment to therapeutic development is the lack of high-fidelity models that account for the fibrotic microenvironment. Our goal is to create a three-dimensional (3D) platform to enable lung fibrosis studies and recapitulate IPF tissue features. We demonstrate that normal lung fibroblasts encapsulated in collagen microspheres can be pushed toward an activated phenotype, treated with FDA-approved therapies, and their fibrotic function quantified using imaging assays (extracellular matrix deposition, contractile protein expression, and microenvironment compaction). Highlighting the system's utility, we further show that fibroblasts isolated from IPF patient lungs maintain fibrotic phenotypes and manifest reduced fibrotic function when treated with epigenetic modifiers. Our system enables enhanced screening due to improved predictability and fidelity compared to 2D systems combined with superior tractability and throughput compared to 3D systems.
Collapse
Affiliation(s)
- Katherine A. Cummins
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, USA
| | - Peter B. Bitterman
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, USA
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
103
|
Boghdady CM, Kalashnikov N, Mok S, McCaffrey L, Moraes C. Revisiting tissue tensegrity: Biomaterial-based approaches to measure forces across length scales. APL Bioeng 2021; 5:041501. [PMID: 34632250 PMCID: PMC8487350 DOI: 10.1063/5.0046093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-generated forces play a foundational role in tissue dynamics and homeostasis and are critically important in several biological processes, including cell migration, wound healing, morphogenesis, and cancer metastasis. Quantifying such forces in vivo is technically challenging and requires novel strategies that capture mechanical information across molecular, cellular, and tissue length scales, while allowing these studies to be performed in physiologically realistic biological models. Advanced biomaterials can be designed to non-destructively measure these stresses in vitro, and here, we review mechanical characterizations and force-sensing biomaterial-based technologies to provide insight into the mechanical nature of tissue processes. We specifically and uniquely focus on the use of these techniques to identify characteristics of cell and tissue "tensegrity:" the hierarchical and modular interplay between tension and compression that provide biological tissues with remarkable mechanical properties and behaviors. Based on these observed patterns, we highlight and discuss the emerging role of tensegrity at multiple length scales in tissue dynamics from homeostasis, to morphogenesis, to pathological dysfunction.
Collapse
Affiliation(s)
| | - Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | - Stephanie Mok
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | | | | |
Collapse
|
104
|
Linde-Medina M, Smit TH. Molecular and Mechanical Cues for Somite Periodicity. Front Cell Dev Biol 2021; 9:753446. [PMID: 34901002 PMCID: PMC8663771 DOI: 10.3389/fcell.2021.753446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Somitogenesis refers to the segmentation of the paraxial mesoderm, a tissue located on the back of the embryo, into regularly spaced and sized pieces, i.e., the somites. This periodicity is important to assure, for example, the formation of a functional vertebral column. Prevailing models of somitogenesis are based on the existence of a gene regulatory network capable of generating a striped pattern of gene expression, which is subsequently translated into periodic tissue boundaries. An alternative view is that the pre-pattern that guides somitogenesis is not chemical, but of a mechanical origin. A striped pattern of mechanical strain can be formed in physically connected tissues expanding at different rates, as it occurs in the embryo. Here we argue that both molecular and mechanical cues could drive somite periodicity and suggest how they could be integrated.
Collapse
Affiliation(s)
| | - Theodoor H. Smit
- Department of Orthopaedic Surgery, Amsterdam Movement Sciences, Amsterdam University Medical Centres, Amsterdam, Netherlands
- Department of Medical Biology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| |
Collapse
|
105
|
Nogueira LFB, Maniglia BC, Buchet R, Millán JL, Ciancaglini P, Bottini M, Ramos AP. Three-dimensional cell-laden collagen scaffolds: From biochemistry to bone bioengineering. J Biomed Mater Res B Appl Biomater 2021; 110:967-983. [PMID: 34793621 DOI: 10.1002/jbm.b.34967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/05/2021] [Accepted: 10/30/2021] [Indexed: 12/22/2022]
Abstract
The bones can be viewed as both an organ and a material. As an organ, the bones give structure to the body, facilitate skeletal movement, and provide protection to internal organs. As a material, the bones consist of a hybrid organic/inorganic three-dimensional (3D) matrix, composed mainly of collagen, noncollagenous proteins, and a calcium phosphate mineral phase, which is formed and regulated by the orchestrated action of a complex array of cells including chondrocytes, osteoblasts, osteocytes, and osteoclasts. The interactions between cells, proteins, and minerals are essential for the bone functions under physiological loading conditions, trauma, and fractures. The organization of the bone's organic and inorganic phases stands out for its mechanical and biological properties and has inspired materials research. The objective of this review is to fill the gaps between the physical and biological characteristics that must be achieved to fabricate scaffolds for bone tissue engineering with enhanced performance. We describe the organization of bone tissue highlighting the characteristics that have inspired the development of 3D cell-laden collagenous scaffolds aimed at replicating the mechanical and biological properties of bone after implantation. The role of noncollagenous macromolecules in the organization of the collagenous matrix and mineralization ability of entrapped cells has also been reviewed. Understanding the modulation of cell activity by the extracellular matrix will ultimately help to improve the biological performance of 3D cell-laden collagenous scaffolds used for bone regeneration and repair as well as for in vitro studies aimed at unravelling physiological and pathological processes occurring in the bone.
Collapse
Affiliation(s)
- Lucas Fabricio Bahia Nogueira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil.,Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Bianca C Maniglia
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| | - Rene Buchet
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ana Paula Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| |
Collapse
|
106
|
A Cylindrical Molding Method for the Biofabrication of Plane-Shaped Skeletal Muscle Tissue. MICROMACHINES 2021; 12:mi12111411. [PMID: 34832821 PMCID: PMC8625600 DOI: 10.3390/mi12111411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/13/2021] [Accepted: 11/14/2021] [Indexed: 11/17/2022]
Abstract
Muscle tissues can be fabricated in vitro by culturing myoblast-populated hydrogels. To counter the shrinkage of the myoblast-populated hydrogels during culture, a pair of anchors are generally utilized to fix the two ends of the hydrogel. Here, we propose an alternative method to counter the shrinkage of the hydrogel and fabricate plane-shaped skeletal muscle tissues. The method forms myoblast-populated hydrogel in a cylindrical cavity with a central pillar, which can prevent tissue shrinkage along the circumferential direction. By eliminating the usages of the anchor pairs, our proposed method can produce plane-shaped skeletal muscle tissues with uniform width and thickness. In experiments, we demonstrate the fabrication of plane-shaped (length: ca. 10 mm, width: 5~15 mm) skeletal muscle tissue with submillimeter thickness. The tissues have uniform shapes and are populated with differentiated muscle cells stained positive for myogenic differentiation markers (i.e., myosin heavy chains). In addition, we show the assembly of subcentimeter-order tissue blocks by stacking the plane-shaped skeletal muscle tissues. The proposed method can be further optimized and scaled up to produce cultured animal products such as cultured meat.
Collapse
|
107
|
Pitta Kruize C, Panahkhahi S, Putra NE, Diaz-Payno P, van Osch G, Zadpoor AA, Mirzaali MJ. Biomimetic Approaches for the Design and Fabrication of Bone-to-Soft Tissue Interfaces. ACS Biomater Sci Eng 2021. [PMID: 34784181 DOI: 10.1021/acsbiomaterials.1c00620] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bone-to-soft tissue interfaces are responsible for transferring loads between tissues with significantly dissimilar material properties. The examples of connective soft tissues are ligaments, tendons, and cartilages. Such natural tissue interfaces have unique microstructural properties and characteristics which avoid the abrupt transitions between two tissues and prevent formation of stress concentration at their connections. Here, we review some of the important characteristics of these natural interfaces. The native bone-to-soft tissue interfaces consist of several hierarchical levels which are formed in a highly specialized anisotropic fashion and are composed of different types of heterogeneously distributed cells. The characteristics of a natural interface can rely on two main design principles, namely by changing the local microarchitectural features (e.g., complex cell arrangements, and introducing interlocking mechanisms at the interfaces through various geometrical designs) and changing the local chemical compositions (e.g., a smooth and gradual transition in the level of mineralization). Implementing such design principles appears to be a promising approach that can be used in the design, reconstruction, and regeneration of engineered biomimetic tissue interfaces. Furthermore, prominent fabrication techniques such as additive manufacturing (AM) including 3D printing and electrospinning can be used to ease these implementation processes. Biomimetic interfaces have several biological applications, for example, to create synthetic scaffolds for osteochondral tissue repair.
Collapse
Affiliation(s)
- Carlos Pitta Kruize
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Sara Panahkhahi
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Niko Eka Putra
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Pedro Diaz-Payno
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Gerjo van Osch
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Mohammad J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
| |
Collapse
|
108
|
Physically-based structural modeling of a typical regenerative tissue analog bridges material macroscale continuum and cellular microscale discreteness and elucidates the hierarchical characteristics of cell-matrix interaction. J Mech Behav Biomed Mater 2021; 126:104956. [PMID: 34930707 DOI: 10.1016/j.jmbbm.2021.104956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 11/01/2021] [Indexed: 11/20/2022]
Abstract
This paper presents a comprehensive physically-based structural modelling for the passive and active biomechanical processes in a typical engineered tissue - namely, cell-compacted collagen gel. First, it introduces a sinusoidal curve analog for quantifying the mechanical response of the collagen fibrils and a probability distribution function of the characteristic crimp ratio for taking into account the fibrillar geometric entropic effect. The constitutive framework based on these structural characteristics precisely reproduces the nonlinearity, the viscoelasticity, and fairly captures the Poisson effect exhibiting in the macroscale tensile tests; which, therefore, substantially validates the structural modelling for the analysis of the cell-gel interaction during collagen gel compaction. Second, a deterministic molecular clutch model specific to the interaction between the cell pseudopodium and the collagen network is developed, which emphasizes the dependence of traction force on clutch number altering with the retrograde flow velocity, actin polymeric velocity, and the deformation of the stretched fibril. The modelling reveals the hierarchical features of cellular substrate sensing, i.e. a biphasic traction force response to substrate elasticity begins at the level of individual fibrils and develops into the second biphasic sensing by means of the fibrillar number integration at the whole-cell level. Singular in crossing the realms of continuum and discrete mechanics, the methodologies developed in this study for modelling the filamentous materials and cell-fibril interaction deliver deep insight into the temporospatially dynamic 3D cell-matrix interaction, and are able to bridge the cellular microscale and material macroscale in the exploration of related topics in mechanobiology.
Collapse
|
109
|
The proportion of the key components analysed in collagen-based isolates from fish and mammalian tissues processed by different protocols. J Food Compost Anal 2021. [DOI: 10.1016/j.jfca.2021.104059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
110
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
111
|
Shaping collagen for engineering hard tissues: Towards a printomics approach. Acta Biomater 2021; 131:41-61. [PMID: 34192571 DOI: 10.1016/j.actbio.2021.06.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022]
Abstract
Hard tissue engineering has evolved over the past decades, with multiple approaches being explored and developed. Despite the rapid development and success of advanced 3D cell culture, 3D printing technologies and material developments, a gold standard approach to engineering and regenerating hard tissue substitutes such as bone, dentin and cementum, has not yet been realised. One such strategy that differs from conventional regenerative medicine approach of other tissues, is the in vitro mineralisation of collagen templates in the absence of cells. Collagen is the most abundant protein within the human body and forms the basis of all hard tissues. Once mineralised, collagen provides important support and protection to humans, for example in the case of bone tissue. Multiple in vitro fabrication strategies and mineralisation approaches have been developed and their success in facilitating mineral deposition on collagen to achieve bone-like scaffolds evaluated. Critical to the success of such fabrication and biomineralisation approaches is the collagen template, and its chemical composition, organisation, and density. The key factors that influence such properties are the collagen processing and fabrication techniques utilised to create the template, and the mineralisation strategy employed to deposit mineral on and throughout the templates. However, despite its importance, relatively little attention has been placed on these two critical factors. Here, we critically examine the processing, fabrication and mineralisation strategies that have been used to mineralise collagen templates, and offer insights and perspectives on the most promising strategies for creating mineralised collagen scaffolds. STATEMENT OF SIGNIFICANCE: In this review, we highlight the critical need to fabricate collagen templates with advanced processing techniques, in a manner that achieves biomimicry of the hierarchical collagen structure, prior to utilising in vitro mineralisation strategies. To this end, we focus on the initial collagen that is selected, the extraction techniques used and the native fibril forming potential retained to create reconstituted collagen scaffolds. This review synthesises current best practises in material sourcing, processing, mineralisation strategies and fabrication techniques, and offers insights into how these can best be exploited in future studies to successfully mineralise collagen templates.
Collapse
|
112
|
Ebhodaghe SO. Natural Polymeric Scaffolds for Tissue Engineering Applications. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:2144-2194. [PMID: 34328068 DOI: 10.1080/09205063.2021.1958185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Natural polymeric scaffolds can be used for tissue engineering applications such as cell delivery and cell-free supporting of native tissues. This is because of their desirable properties such as; high biocompatibility, tunable mechanical strength and conductivity, large surface area, porous- and extracellular matrix (ECM)-mimicked structures. Specifically, their less toxicity and biocompatibility makes them suitable for several tissue engineering applications. For these reasons, several biopolymeric scaffolds are currently being explored for numerous tissue engineering applications. To date, research on the nature, chemistry, and properties of nanocomposite biopolymers are been reported, while the need for a comprehensive research note on more tissue engineering application of these biopolymers remains. As a result, this present study comprehensively reviews the development of common natural biopolymers as scaffolds for tissue engineering applications such as cartilage tissue engineering, cornea repairs, osteochondral defect repairs, and nerve regeneration. More so, the implications of research findings for further studies are presented, while the impact of research advances on future research and other specific recommendations are added as well.
Collapse
|
113
|
Yu Z, Liu KK. Soft Polymer-Based Technique for Cellular Force Sensing. Polymers (Basel) 2021; 13:2672. [PMID: 34451211 PMCID: PMC8399510 DOI: 10.3390/polym13162672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 01/03/2023] Open
Abstract
Soft polymers have emerged as a vital type of material adopted in biomedical engineering to perform various biomechanical characterisations such as sensing cellular forces. Distinct advantages of these materials used in cellular force sensing include maintaining normal functions of cells, resembling in vivo mechanical characteristics, and adapting to the customised functionality demanded in individual applications. A wide range of techniques has been developed with various designs and fabrication processes for the desired soft polymeric structures, as well as measurement methodologies in sensing cellular forces. This review highlights the merits and demerits of these soft polymer-based techniques for measuring cellular contraction force with emphasis on their quantitativeness and cell-friendliness. Moreover, how the viscoelastic properties of soft polymers influence the force measurement is addressed. More importantly, the future trends and advancements of soft polymer-based techniques, such as new designs and fabrication processes for cellular force sensing, are also addressed in this review.
Collapse
Affiliation(s)
| | - Kuo-Kang Liu
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK;
| |
Collapse
|
114
|
Sun Y, Xu H, Xu X, Wang H, Yuan Y, An Z, Xu Z, Wang G. A novel method to obtain rat aortic media for primary culture of rat aortic smooth muscle cells. In Vitro Cell Dev Biol Anim 2021; 57:726-734. [PMID: 34462813 DOI: 10.1007/s11626-021-00615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/10/2021] [Indexed: 11/27/2022]
Abstract
An efficient and simple method to obtain aortic media for primary culture of rat vascular smooth muscle cells (RVSMCs) is developed. The main steps to obtain aortic media include isolation of rat aortic artery, removal of the fat tissue and branches, separation of longitudinal cutting edge, and peeling off the adventitia. Then, aortic media was used to obtain RVSMCs by our tissue explants method and the enzyme digestion method. The removal efficiency of the intima and adventitia was confirmed by hematoxylin-eosin and immunohistochemical staining. Morphology and immunofluorescent staining were used to identify cells and cell purity. RVSMCs at the 3rd and 8th passages were isolated by our tissue explants method; the enzyme digestion method and the traditional tissue explants method were compared respectively. Western blotting and gel contraction assay were used to investigate the phenotype and contraction ability of RVSMCs obtained by the different methods. Compared with the other methods, RVSMCs isolated by our method showed higher purity and demonstrated "contractile" phenotype with retained contraction ability for more passages. And the aortic media obtained showed no visible damage with few endothelial cells and fibroblasts remained. An efficient and simple method was established to obtain rat aortic media for primary culture of RVSMCs with high purity, "contractile" phenotype characteristics, and more stable during subculturing.
Collapse
Affiliation(s)
- Yangyong Sun
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Hongjie Xu
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiangyang Xu
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - He Wang
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ye Yuan
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhao An
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Guokun Wang
- Department of Cardiovascular Surgery, Changhai Hospital, The Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
115
|
Ramírez OJ, Alvarez S, Contreras-Kallens P, Barrera NP, Aguayo S, Schuh CMAP. Type I collagen hydrogels as a delivery matrix for royal jelly derived extracellular vesicles. Drug Deliv 2021; 27:1308-1318. [PMID: 32924637 PMCID: PMC7534280 DOI: 10.1080/10717544.2020.1818880] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Throughout the last decade, extracellular vesicles (EVs) have become increasingly popular
in several areas of regenerative medicine. Recently, Apis
mellifera royal jelly EVs (RJ EVs) were shown to display favorable wound
healing properties such as stimulation of mesenchymal stem cell migration and inhibition
of staphylococcal biofilms. However, the sustained and effective local delivery of EVs in
non-systemic approaches – such as patches for chronic cutaneous wounds – remains an
important challenge for the development of novel EV-based wound healing therapies.
Therefore, the present study aimed to assess the suitability of type I collagen -a
well-established biomaterial for wound healing – as a continuous delivery matrix. RJ EVs
were integrated into collagen gels at different concentrations, where gels containing
2 mg/ml collagen were found to display the most stable release kinetics. Functionality of
released RJ EVs was confirmed by assessing fibroblast EV uptake and migration in a wound
healing assay. We could demonstrate reliable EV uptake into fibroblasts with a sustained
pro-migratory effect for up to 7 d. Integrating fibroblasts into the RJ EV-containing
collagen gel increased the contractile capacity of these cells, confirming availability of
RJ EVs to fibroblasts within the collagen gel. Furthermore, EVs released from collagen
gels were found to inhibit Staphylococcus aureus ATCC 29213
biofilm formation. Overall, our results suggest that type I collagen could be utilized as
a reliable, reproducible release system to deliver functional RJ EVs for wound healing
therapies.
Collapse
Affiliation(s)
- Orlando J Ramírez
- Facultad de Medicina, Centro de Medicina Regenerativa, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Simón Alvarez
- Facultad de Medicina, Centro de Medicina Regenerativa, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Pamina Contreras-Kallens
- Facultad de Medicina, Centro de Medicina Regenerativa, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Nelson P Barrera
- Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastian Aguayo
- Faculty of Medicine, Dentistry School, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christina M A P Schuh
- Facultad de Medicina, Centro de Medicina Regenerativa, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
116
|
Fournier R, Harrison RE. Methods for studying MLO-Y4 osteocytes in collagen-hydroxyapatite scaffolds in the rotary cell culture system. Connect Tissue Res 2021; 62:436-453. [PMID: 32375524 DOI: 10.1080/03008207.2020.1764548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: The rotary cell culture system (RCCS) is a common clinorotation device for cell culture. It is also used as a low-shear suspension culture bioreactor to form functionalized 3D tissue constructs and to model microgravity. We sought to develop a 3D scaffold composed of type I collagen and hydroxyapatite (collagen-HA) to characterize MLO-Y4 osteocytes following suspension culture or clinorotation.Materials and Methods: MLO-Y4 cells were embedded in collagen-HA. The scaffold was formed into droplets for suspension culture or wall-adhered to the RCCS for clinorotation. AFM, rheometry, immunofluorescence and qRT-PCR were employed to measure the scaffold stiffness, cell viability and gene expression of cells in collagen-HA scaffolds. Dendritic cells were visualized and quantified and gene expression after suspension culture and clinorotation was compared to static controls.Results: The optimized scaffold for the RCCS consisted of collagen with 6 mg/mL HA which had a stiffness of < 1 kPa. MLO-Y4 cell viability was higher in collagen-HA scaffolds, compared to scaffolds without HA. Collagen-HA scaffolds induced higher osteocyte-specific gene expression compared to cells cultured on 2D plastic. Cells in the scaffold downregulated DMP1, E11, IL-6, and RANKL, and had fewer dendritic cells following suspension culture whereas clinorotation downregulated DMP1 and E11 genes, compared to static controls.Conclusions: Suspension culture for 3 days in collagen-HA stimulates growth of osteocytes but may also desensitize them to mechanical cues. Clinorotation for 3 days in collagen-HA does not stimulate proliferation or expression of mechanosensitive genes, indicating that it may be an effective mechanical unloading environment.
Collapse
Affiliation(s)
- Roxanne Fournier
- Department of Cell & Systems Biology and the Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Rene E Harrison
- Department of Cell & Systems Biology and the Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
117
|
Sun Y, Myers DR, Nikolov SV, Oshinowo O, Baek J, Bowie SM, Lambert TP, Woods E, Sakurai Y, Lam WA, Alexeev A. Platelet heterogeneity enhances blood clot volumetric contraction: An example of asynchrono-mechanical amplification. Biomaterials 2021; 274:120828. [PMID: 33964792 PMCID: PMC8184644 DOI: 10.1016/j.biomaterials.2021.120828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 04/01/2021] [Accepted: 04/11/2021] [Indexed: 01/22/2023]
Abstract
Physiological processes such as blood clotting and wound healing as well as pathologies such as fibroses and musculoskeletal contractures, all involve biological materials composed of a contracting cellular population within a fibrous matrix, yet how the microscale interactions among the cells and the matrix lead to the resultant emergent behavior at the macroscale tissue level remains poorly understood. Platelets, the anucleate cell fragments that do not divide nor synthesize extracellular matrix, represent an ideal model to study such systems. During blood clot contraction, microscopic platelets actively pull fibers to shrink the macroscale clot to less than 10% of its initial volume. We discovered that platelets utilize a new emergent behavior, asynchrono-mechanical amplification, to enhanced volumetric material contraction and to magnify contractile forces. This behavior is triggered by the heterogeneity in the timing of a population of actuators. This result indicates that cell heterogeneity, often attributed to stochastic cell-to-cell variability, can carry an essential biophysical function, thereby highlighting the importance of considering 4 dimensions (space + time) in cell-matrix biomaterials. This concept of amplification via heterogeneity can be harnessed to increase mechanical efficiency in diverse systems including implantable biomaterials, swarm robotics, and active polymer composites.
Collapse
Affiliation(s)
- Yueyi Sun
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - David R Myers
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA; The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Svetoslav V Nikolov
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - Oluwamayokun Oshinowo
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA; The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - John Baek
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA; The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Samuel M Bowie
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - Tamara P Lambert
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Eric Woods
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Yumiko Sakurai
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA; The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Wilbur A Lam
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA; The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| | - Alexander Alexeev
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA.
| |
Collapse
|
118
|
Oyama TG, Oyama K, Kimura A, Yoshida F, Ishida R, Yamazaki M, Miyoshi H, Taguchi M. Collagen hydrogels with controllable combined cues of elasticity and topography to regulate cellular processes. Biomed Mater 2021; 16. [PMID: 34030146 DOI: 10.1088/1748-605x/ac0452] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
The elasticity, topography, and chemical composition of cell culture substrates influence cell behavior. However, the cellular responses toin vivoextracellular matrix (ECM), a hydrogel of proteins (mainly collagen) and polysaccharides, remain unknown as there is no substrate that preserves the key features of native ECM. This study introduces novel collagen hydrogels that can combine elasticity, topography, and composition and reproduce the correlation between collagen concentration (C) and elastic modulus (E) in native ECM. A simple reagent-free method based on radiation-cross-linking altered ECM-derived collagen I and hydrolyzed collagen (gelatin or collagen peptide) solutions into hydrogels with tunable elastic moduli covering a broad range of soft tissues (E= 1-236 kPa) originating from the final collagen density in the hydrogels (C= 0.3%-14%) and precise microtopographies (⩾1 μm). The amino acid composition ratio was almost unchanged by this method, and the obtained collagen hydrogels maintained enzyme-mediated degradability. These collagen hydrogels enabled investigation of the responses of cell lines (fibroblasts, epithelial cells, and myoblasts) and primary cells (rat cardiomyocytes) to soft topographic cues such as thosein vivounder the positive correlation betweenCandE. These cells adhered directly to the collagen hydrogels and chose to stay atop or spontaneously migrate into them depending onE, that is, the density of the collagen network,C. We revealed that the cell morphology and actin cytoskeleton organization conformed to the topographic cues, even when they are as soft asin vivoECM. The stiffer microgrooves on collagen hydrogels aligned cells more effectively, except HeLa cells that underwent drastic changes in cell morphology. These collagen hydrogels may not only reducein vivoandin vitrocell behavioral disparity but also facilitate artificial ECM design to control cell function and fate for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tomoko G Oyama
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan
| | - Kotaro Oyama
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan.,PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Atsushi Kimura
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan
| | - Fumiya Yoshida
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan.,Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma 376-0052, Japan
| | - Ryo Ishida
- Graduate School of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Masashi Yamazaki
- Graduate School of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Hiromi Miyoshi
- Graduate School of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Mitsumasa Taguchi
- Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 1233 Watanukimachi, Takasaki-shi, Gunma 370-1292, Japan
| |
Collapse
|
119
|
Hertig D, Maddah S, Memedovski R, Kurth S, Moreno A, Pennestri M, Felser A, Nuoffer JM, Vermathen P. Live monitoring of cellular metabolism and mitochondrial respiration in 3D cell culture system using NMR spectroscopy. Analyst 2021; 146:4326-4339. [PMID: 34106111 PMCID: PMC8239994 DOI: 10.1039/d1an00041a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Because of the interplay between mitochondrial respiration and cellular metabolism, the simultaneous monitoring of both cellular processes provides important insights for the understanding of biological processes. NMR flow systems provide a unique window into the metabolome of cultured cells. Simplified bioreactor construction based on commercially available flow systems increase the practicability and reproducibility of bioreactor studies using standard NMR spectrometers. We therefore aim at establishing a reproducible NMR bioreactor system for metabolic 1H-NMR investigations of small molecules and concurrent oxygenation determination by 19F-NMR, with in depth description and validation by accompanying measures. Methods: We demonstrate a detailed and standardized workflow for the preparation and transfer of collagen based 3D cell culture of high cell density for perfused investigation in a 5 mm NMR tube. Self-constructed gas mixing station enables 5% CO2 atmosphere for physiological pH in carbon based medium and is perfused by HPLC pump. Results & Discussion: Implemented perfused bioreactor allows detection of perfusion rate dependent metabolite content. We show interleaved dynamic profiling of 26 metabolites and mitochondrial respiration. During constant perfusion, sequential injection of rotenone/oligomycin and 2-deoxy-glucose indicated immediate activation and deactivation of glycolytic rate and full inhibition of oxygen consumption. We show sensitivity to detect substrate degradation rates of major mitochondrial fuel pathways and were able to simultaneously measure cellular oxygen consumption. We show sensitivity to detect substrate degradation rates of major mitochondrial fuel pathways and feasibility to simultaneously measure cellular oxygen consumption combining a commercially available flow tube system with a standard 5 mm NMR probe.![]()
Collapse
Affiliation(s)
- Damian Hertig
- Department of Biomedical Research and Radiology, University of Bern, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Menon SN, Flegg JA. Mathematical Modeling Can Advance Wound Healing Research. Adv Wound Care (New Rochelle) 2021; 10:328-344. [PMID: 32634070 PMCID: PMC8082733 DOI: 10.1089/wound.2019.1132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 06/26/2020] [Indexed: 12/27/2022] Open
Abstract
Significance: For over 30 years, there has been sustained interest in the development of mathematical models for investigating the complex mechanisms underlying each stage of the wound healing process. Despite the immense associated challenges, such models have helped usher in a paradigm shift in wound healing research. Recent Advances: In this article, we review contributions in the field that span epidermal, dermal, and corneal wound healing, and treatments of nonhealing wounds. The recent influence of mathematical models on biological experiments is detailed, with a focus on wound healing assays and fibroblast-populated collagen lattices. Critical Issues: We provide an overview of the field of mathematical modeling of wound healing, highlighting key advances made in recent decades, and discuss how such models have contributed to the development of improved treatment strategies and/or an enhanced understanding of the tightly regulated steps that comprise the healing process. Future Directions: We detail some of the open problems in the field that could be addressed through a combination of theoretical and/or experimental approaches. To move the field forward, we need to have a common language between scientists to facilitate cross-collaboration, which we hope this review can support by highlighting progress to date.
Collapse
Affiliation(s)
| | - Jennifer A. Flegg
- School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
121
|
Eichinger JF, Haeusel LJ, Paukner D, Aydin RC, Humphrey JD, Cyron CJ. Mechanical homeostasis in tissue equivalents: a review. Biomech Model Mechanobiol 2021; 20:833-850. [PMID: 33683513 PMCID: PMC8154823 DOI: 10.1007/s10237-021-01433-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/04/2021] [Indexed: 12/20/2022]
Abstract
There is substantial evidence that growth and remodeling of load bearing soft biological tissues is to a large extent controlled by mechanical factors. Mechanical homeostasis, which describes the natural tendency of such tissues to establish, maintain, or restore a preferred mechanical state, is thought to be one mechanism by which such control is achieved across multiple scales. Yet, many questions remain regarding what promotes or prevents homeostasis. Tissue equivalents, such as collagen gels seeded with living cells, have become an important tool to address these open questions under well-defined, though limited, conditions. This article briefly reviews the current state of research in this area. It summarizes, categorizes, and compares experimental observations from the literature that focus on the development of tension in tissue equivalents. It focuses primarily on uniaxial and biaxial experimental studies, which are well-suited for quantifying interactions between mechanics and biology. The article concludes with a brief discussion of key questions for future research in this field.
Collapse
Affiliation(s)
- Jonas F Eichinger
- Institute for Computational Mechanics, Technical University of Munich, 85748, Munich, Germany
- Institute of Continuum and Materials Mechanics, Hamburg University of Technology, 21073, Hamburg, Germany
| | - Lea J Haeusel
- Institute for Computational Mechanics, Technical University of Munich, 85748, Munich, Germany
| | - Daniel Paukner
- Institute of Continuum and Materials Mechanics, Hamburg University of Technology, 21073, Hamburg, Germany
- Institute of Material Systems Modeling, Helmholtz-Zentrum Geesthacht, 21502, Geesthacht, Germany
| | - Roland C Aydin
- Institute of Material Systems Modeling, Helmholtz-Zentrum Geesthacht, 21502, Geesthacht, Germany
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Christian J Cyron
- Institute of Continuum and Materials Mechanics, Hamburg University of Technology, 21073, Hamburg, Germany.
- Institute of Material Systems Modeling, Helmholtz-Zentrum Geesthacht, 21502, Geesthacht, Germany.
| |
Collapse
|
122
|
Mechanical and Immunological Regulation in Wound Healing and Skin Reconstruction. Int J Mol Sci 2021; 22:ijms22115474. [PMID: 34067386 PMCID: PMC8197020 DOI: 10.3390/ijms22115474] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
In the past decade, a new frontier in scarless wound healing has arisen because of significant advances in the field of wound healing realised by incorporating emerging concepts from mechanobiology and immunology. The complete integumentary organ system (IOS) regeneration and scarless wound healing mechanism, which occurs in specific species, body sites and developmental stages, clearly shows that mechanical stress signals and immune responses play important roles in determining the wound healing mode. Advances in tissue engineering technology have led to the production of novel human skin equivalents and organoids that reproduce cell–cell interactions with tissue-scale tensional homeostasis, and enable us to evaluate skin tissue morphology, functionality, drug response and wound healing. This breakthrough in tissue engineering has the potential to accelerate the understanding of wound healing control mechanisms through complex mechanobiological and immunological interactions. In this review, we present an overview of recent studies of biomechanical and immunological wound healing and tissue remodelling mechanisms through comparisons of species- and developmental stage-dependent wound healing mechanisms. We also discuss the possibility of elucidating the control mechanism of wound healing involving mechanobiological and immunological interaction by using next-generation human skin equivalents.
Collapse
|
123
|
Salameh S, Tissot N, Cache K, Lima J, Suzuki I, Marinho PA, Rielland M, Soeur J, Takeuchi S, Germain S, Breton L. A perfusable vascularized full-thickness skin model for potential topical and systemic applications. Biofabrication 2021; 13. [PMID: 33910175 DOI: 10.1088/1758-5090/abfca8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/28/2021] [Indexed: 12/12/2022]
Abstract
Vascularization of reconstructed tissues is one of the remaining hurdles to be considered to improve both the functionality and viability of skin grafts and the relevance ofin vitroapplications. Our study, therefore, sought to develop a perfusable vascularized full-thickness skin equivalent that comprises a more complex blood vasculature compared to existing models. We combined molding, auto-assembly and microfluidics techniques in order to create a vascularized skin equivalent representing (a) a differentiated epidermis with a physiological organization and correctly expressing K14, K10, Involucrin, TGM1 and Filaggrin, (b) three perfusable vascular channels with angiogenic sprouts stained with VE-Caderin and Collagen IV, (c) an adjacent microvascular network created via vasculogenesis and connected to the sprouting macrovessels. Histological analysis and immunostaining of CD31, Collagen IV, Perlecan and Laminin proved the integrity of vascular constructs. In order to validate the vascularized skin potential of topical and systemic applications, caffeine and minoxidil, two compounds with different chemical properties, were topically applied to measure skin permeability and benzo[a]pyrene pollutant was systemically applied to evaluate systemic delivery. Our results demonstrated that perfusion of skin reconstructs and the presence of a complex vascular plexus resulted in a more predictive and reliable model to assess respectively topical and systemic applications. This model is therefore aimed at furthering drug discovery and improving clinical translation in dermatology.
Collapse
Affiliation(s)
- Sacha Salameh
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France.,Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Nicolas Tissot
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Kevin Cache
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Joaquim Lima
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Itaru Suzuki
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | | | - Maité Rielland
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Jérémie Soeur
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Shoji Takeuchi
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Lionel Breton
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| |
Collapse
|
124
|
Pokki J, Zisi I, Schulman E, Indana D, Chaudhuri O. Magnetic probe-based microrheology reveals local softening and stiffening of 3D collagen matrices by fibroblasts. Biomed Microdevices 2021; 23:27. [PMID: 33900463 PMCID: PMC8076128 DOI: 10.1007/s10544-021-00547-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Changes in extracellular matrix stiffness impact a variety of biological processes including cancer progression. However, cells also actively remodel the matrices they interact with, dynamically altering the matrix mechanics they respond to. Further, cells not only react to matrix stiffness, but also have a distinct reaction to matrix viscoelasticity. The impact of cell-driven matrix remodeling on matrix stiffness and viscoelasticity at the microscale remains unclear, as existing methods to measure mechanics are largely at the bulk scale or probe only the surface of matrices, and focus on stiffness. Yet, establishing the impact of the matrix remodeling at the microscale is crucial to obtaining an understanding of mechanotransduction in biological matrices, and biological matrices are not just elastic, but are viscoelastic. Here, we advanced magnetic probe-based microrheology to overcome its previous limitations in measuring viscoelasticity at the cell-size-scale spatial resolution within 3D cell cultures that have tissue-relevant stiffness levels up to a Young's modulus of 0.5 kPa. Our magnetic microrheometers exert controlled magnetic forces on magnetic microprobes within reconstituted extracellular matrices and detect microprobe displacement responses to measure matrix viscoelasticity and determine the frequency-dependent shear modulus (stiffness), the loss tangent, and spatial heterogeneity. We applied these tools to investigate how microscale viscoelasticity of collagen matrices is altered by fibroblast cells as they contract collagen gels, a process studied extensively at the macroscale. Interestingly, we found that fibroblasts first soften the matrix locally over the first 32 hours of culture, and then progressively stiffen the matrix thereafter. Fibroblast activity also progressively increased the matrix loss tangent. We confirmed that the softening is caused by matrix-metalloproteinase-mediated collagen degradation, whereas stiffening is associated with local alignment and densification of collagen fibers around the fibroblasts. This work paves the way for the use of measurement systems that quantify microscale viscoelasticity within 3D cell cultures for studies of cell-matrix interactions in cancer progression and other areas.
Collapse
Affiliation(s)
- Juho Pokki
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA. .,Department of Electrical Engineering and Automation, Aalto University, Espoo, Finland.
| | - Iliana Zisi
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ester Schulman
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
125
|
Allbritton-King JD, Kimicata M, Fisher JP. Incorporating a structural extracellular matrix gradient into a porcine urinary bladder matrix-based hydrogel dermal scaffold. J Biomed Mater Res A 2021; 109:1893-1904. [PMID: 33797180 DOI: 10.1002/jbm.a.37181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/17/2021] [Accepted: 03/24/2021] [Indexed: 12/21/2022]
Abstract
The increasing prevalence of chronic, nonhealing wounds necessitates the investigation of full-thickness skin substitutes conducive to host integration and wound closure. Extracellular matrix (ECM)-based hydrogel scaffolds mimic the physiological matrix environment of dermal cells, thereby conferring favorable cellular adhesion, infiltration, and proliferation. However, low-concentration ECM hydrogels rapidly lose mechanical strength as they degrade, leaving them susceptible to shrinkage from fibroblast-mediated contraction. Conversely, high-concentration ECM hydrogels are typically too dense to permit nutrient diffusion and cellular migration. This study investigates the design and fabrication of a graded-concentration hydrogel composed of porcine urinary bladder matrix (UBM) as a dermal scaffold for potential use in chronic wound treatment. Our method of UBM isolation and decellularization effectively removed native DNA while preserving matrix proteins. Hydrogels composed of a range of decellularized UBM (dUBM) concentrations were characterized and used to design a three-tiered gradient hydrogel that promoted cellular activity and maintained structural integrity. The gradient dUBM hydrogel showed stability of cross-sectional area during collagenase degradation, despite considerable loss of mass. The gradient dUBM hydrogel also resisted fibroblast-mediated contraction while supporting high surface cell viability, demonstrating the mechanical support provided by denser layers of dUBM. Overall, incorporation of an ECM concentration gradient into a porcine UBM-based hydrogel scaffold capitalizes on the unique advantages of both high and low-concentration ECM hydrogels, and mitigates the structural weaknesses that have limited the efficacy of hydrogel dermal scaffolds for chronic wounds. Our gradient design shows promise for future development of stable, pro-regenerative wound scaffolds with customized architectures using 3D printing.
Collapse
Affiliation(s)
- Jules D Allbritton-King
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA.,Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Megan Kimicata
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA.,Department of Materials Science and Engineering, University of Maryland, College Park, Maryland, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA.,Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
126
|
Ng AHM, Khoshakhlagh P, Rojo Arias JE, Pasquini G, Wang K, Swiersy A, Shipman SL, Appleton E, Kiaee K, Kohman RE, Vernet A, Dysart M, Leeper K, Saylor W, Huang JY, Graveline A, Taipale J, Hill DE, Vidal M, Melero-Martin JM, Busskamp V, Church GM. A comprehensive library of human transcription factors for cell fate engineering. Nat Biotechnol 2021; 39:510-519. [PMID: 33257861 PMCID: PMC7610615 DOI: 10.1038/s41587-020-0742-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
Human pluripotent stem cells (hPSCs) offer an unprecedented opportunity to model diverse cell types and tissues. To enable systematic exploration of the programming landscape mediated by transcription factors (TFs), we present the Human TFome, a comprehensive library containing 1,564 TF genes and 1,732 TF splice isoforms. By screening the library in three hPSC lines, we discovered 290 TFs, including 241 that were previously unreported, that induce differentiation in 4 days without alteration of external soluble or biomechanical cues. We used four of the hits to program hPSCs into neurons, fibroblasts, oligodendrocytes and vascular endothelial-like cells that have molecular and functional similarity to primary cells. Our cell-autonomous approach enabled parallel programming of hPSCs into multiple cell types simultaneously. We also demonstrated orthogonal programming by including oligodendrocyte-inducible hPSCs with unmodified hPSCs to generate cerebral organoids, which expedited in situ myelination. Large-scale combinatorial screening of the Human TFome will complement other strategies for cell engineering based on developmental biology and computational systems biology.
Collapse
Affiliation(s)
- Alex H M Ng
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- GC Therapeutics, Inc, Cambridge, MA, USA
| | - Parastoo Khoshakhlagh
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- GC Therapeutics, Inc, Cambridge, MA, USA
| | - Jesus Eduardo Rojo Arias
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Giovanni Pasquini
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Anka Swiersy
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Seth L Shipman
- Gladstone Institutes and University of California, San Francisco, San Francisco, CA, USA
| | - Evan Appleton
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- GC Therapeutics, Inc, Cambridge, MA, USA
| | - Kiavash Kiaee
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- GC Therapeutics, Inc, Cambridge, MA, USA
| | - Richie E Kohman
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Andyna Vernet
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Matthew Dysart
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Kathleen Leeper
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Wren Saylor
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Jeremy Y Huang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Amanda Graveline
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Jussi Taipale
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Applied Tumor Genomics Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David E Hill
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marc Vidal
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Volker Busskamp
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany.
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany.
| | - George M Church
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
- GC Therapeutics, Inc, Cambridge, MA, USA.
| |
Collapse
|
127
|
Braun M, Rýglová Š, Suchý T. Determination of glycosaminoglycans in biological matrices using a simple and sensitive reversed-phase HPLC method with fluorescent detection. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1173:122626. [PMID: 33887670 DOI: 10.1016/j.jchromb.2021.122626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 11/25/2022]
Abstract
This paper suggests a sensitive reversed-phase gradient HPLC method combined with fluorescence detection that has been developed, optimized and tested via the quantitative analysis of authentic biological material in an effort to determine and subsequently compare the total content of glycosaminoglycans (GAGs) in various collagen-based biomaterials intended for medical application. The proposed analytical method enabled the identification and separation of the GAGs present from the other components in the samples using commonly-available laboratory equipment; moreover, the very low detection limit of the method permits the determination of GAGs even for very small samples. This study describes the development of the method, including the isolation and processing of the collagen samples prior to HPLC analysis and the optimal parameters applied during the chromatographic analysis. The application of the method in laboratory practice is documented by means of several examples of the determination of GAGs employing both commercial standards and real collagen samples isolated from various animal tissues.
Collapse
Affiliation(s)
- Martin Braun
- Institute of Rock Structure and Mechanics, Czech Academy of Sciences, V Holešovičkách 41, Prague 8 182 09, Czech Republic.
| | - Šárka Rýglová
- Institute of Rock Structure and Mechanics, Czech Academy of Sciences, V Holešovičkách 41, Prague 8 182 09, Czech Republic
| | - Tomáš Suchý
- Institute of Rock Structure and Mechanics, Czech Academy of Sciences, V Holešovičkách 41, Prague 8 182 09, Czech Republic; Faculty of Mechanical Engineering, Czech Technical University in Prague, Technická 4, Prague 6 166 07, Czech Republic
| |
Collapse
|
128
|
Kim K, Kim J, Kim H, Sung GY. Effect of α-Lipoic Acid on the Development of Human Skin Equivalents Using a Pumpless Skin-on-a-Chip Model. Int J Mol Sci 2021; 22:2160. [PMID: 33671528 PMCID: PMC7927099 DOI: 10.3390/ijms22042160] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Owing to the prohibition of cosmetic animal testing, various attempts have recently been made using skin-on-a-chip (SOC) technology as a replacement for animal testing. Previously, we reported the development of a pumpless SOC capable of drug testing with a simple drive using the principle that the medium flows along the channel by gravity when the chip is tilted using a microfluidic channel. In this study, using pumpless SOC, instead of drug testing at the single-cell level, we evaluated the efficacy of α-lipoic acid (ALA), which is known as an anti-aging substance in skin equivalents, for skin tissue and epidermal structure formation. The expression of proteins and changes in genotyping were compared and evaluated. Hematoxylin and eosin staining for histological analysis showed a difference in the activity of fibroblasts in the dermis layer with respect to the presence or absence of ALA. We observed that the epidermis layer became increasingly prominent as the culture period was extended by treatment with 10 μM ALA. The expression of epidermal structural proteins of filaggrin, involucrin, keratin 10, and collagen IV increased because of the effect of ALA. Changes in the epidermis layer were noticeable after the ALA treatment. As a result of aging, damage to the skin-barrier function and structural integrity is reduced, indicating that ALA has an anti-aging effect. We performed a gene analysis of filaggrin, involucrin, keratin 10, integrin, and collagen I genes in ALA-treated human skin equivalents, which indicated an increase in filaggrin gene expression after ALA treatment. These results indicate that pumpless SOC can be used as an in vitro skin model similar to human skin, protein and gene expression can be analyzed, and it can be used for functional drug tests of cosmetic materials in the future. This technology is expected to contribute to the development of skin disease models.
Collapse
Affiliation(s)
- Kyunghee Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (K.K.); (J.K.); (H.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Jisue Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (K.K.); (J.K.); (H.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Hyoungseob Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (K.K.); (J.K.); (H.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (K.K.); (J.K.); (H.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
- Major in Materials Science and Engineering, School of Future Convergence, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
129
|
Signatures of Dermal Fibroblasts from RDEB Pediatric Patients. Int J Mol Sci 2021; 22:ijms22041792. [PMID: 33670258 PMCID: PMC7918539 DOI: 10.3390/ijms22041792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
The recessive form of dystrophic epidermolysis bullosa (RDEB) is a debilitating disease caused by impairments in the junctions of the dermis and the basement membrane of the epidermis. Mutations in the COL7A1 gene induce multiple abnormalities, including chronic inflammation and profibrotic changes in the skin. However, the correlations between the specific mutations in COL7A1 and their phenotypic output remain largely unexplored. The mutations in the COL7A1 gene, described here, were found in the DEB register. Among them, two homozygous mutations and two cases of compound heterozygous mutations were identified. We created the panel of primary patient-specific RDEB fibroblast lines (FEB) and compared it with control fibroblasts from healthy donors (FHC). The set of morphological features and the contraction capacity of the cells distinguished FEB from FHC. We also report the relationships between the mutations and several phenotypic traits of the FEB. Based on the analysis of the available RNA-seq data of RDEB fibroblasts, we performed an RT-qPCR gene expression analysis of our cell lines, confirming the differential status of multiple genes while uncovering the new ones. We anticipate that our panels of cell lines will be useful not only for studying RDEB signatures but also for investigating the overall mechanisms involved in disease progression.
Collapse
|
130
|
Puetzer JL, Ma T, Sallent I, Gelmi A, Stevens MM. Driving Hierarchical Collagen Fiber Formation for Functional Tendon, Ligament, and Meniscus Replacement. Biomaterials 2021; 269:120527. [PMID: 33246739 PMCID: PMC7883218 DOI: 10.1016/j.biomaterials.2020.120527] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/09/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022]
Abstract
Hierarchical collagen fibers are the primary source of strength in musculoskeletal tendons, ligaments, and menisci. It has remained a challenge to develop these large fibers in engineered replacements or in vivo after injury. The objective of this study was to investigate the ability of restrained cell-seeded high density collagen gels to drive hierarchical fiber formation for multiple musculoskeletal tissues. We found boundary conditions applied to high density collagen gels were capable of driving tenocytes, ligament fibroblasts, and meniscal fibrochondrocytes to develop native-sized hierarchical collagen fibers 20-40 μm in diameter. The fibers organize similar to bovine juvenile collagen with native fibril banding patterns and hierarchical fiber bundles 50-350 μm in diameter by 6 weeks. Mirroring fiber organization, tensile properties of restrained samples improved significantly with time, reaching ~1 MPa. Additionally, tendon, ligament, and meniscal cells produced significantly different sized fibers, different degrees of crimp, and different GAG concentrations, which corresponded with respective juvenile tissue. To our knowledge, these are some of the largest, most organized fibers produced to date in vitro. Further, cells produced tissue specific hierarchical fibers, suggesting this system is a promising tool to better understand cellular regulation of fiber formation to better stimulate it in vivo after injury.
Collapse
Affiliation(s)
- Jennifer L Puetzer
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ; Department of Biomedical Engineering and Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States, 23284.
| | - Tianchi Ma
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Ignacio Sallent
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Amy Gelmi
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ.
| |
Collapse
|
131
|
Hartinger JM, Lukáč P, Mitáš P, Mlček M, Popková M, Suchý T, Šupová M, Závora J, Adámková V, Benáková H, Slanař O, Šíma M, Bartoš M, Chlup H, Grus T. Vancomycin-releasing cross-linked collagen sponges as wound dressings. Bosn J Basic Med Sci 2021; 21:61-70. [PMID: 31782696 PMCID: PMC7861629 DOI: 10.17305/bjbms.2019.4496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
The study presents a novel vancomycin-releasing collagen wound dressing derived from Cyprinus carpio collagen type I cross-linked with carbodiimide which retarded the degradation rate and increased the stability of the sponge. Following lyophilization, the dressings were subjected to gamma sterilization. The structure was evaluated via scanning electron microscopy images, micro-computed tomography, and infrared spectrometry. The structural stability and vancomycin release properties were evaluated in phosphate buffered saline. Microbiological testing and a rat model of a wound infected with methicillin-resistant Staphylococcus aureus (MRSA) were then employed to test the efficacy of the treatment of the infected wound. Following an initial mass loss due to the release of vancomycin, the sponges remained stable. After 7 days of exposure in phosphate buffered saline (37°C), 60% of the material remained with a preserved collagen secondary structure together with a high degree of open porosity (over 80%). The analysis of the release of vancomycin revealed homogeneous distribution of the antibiotic both across and between the sponges. The release of vancomycin was retarded as proved by in vitro testing and further confirmed by the animal model from which measurable concentrations were observed in blood samples 24 hours after the subcutaneous implantation of the sponge, which was more than observed following intraperitoneal administration. The sponge was also highly effective in terms of reducing the number of colony-forming units in biopsies extracted from the infected wounds 4 days following the inoculation of the wounds with the MRSA solution. The presented sponges have ideal properties to serve as wound dressing for prevention of surgical site infection or treatment of already infected wounds.
Collapse
Affiliation(s)
- Jan Miroslav Hartinger
- Department of Clinical Pharmacology and Pharmacy, Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Peter Lukáč
- 2nd Department of Cardiovascular Surgery, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petr Mitáš
- 2nd Department of Cardiovascular Surgery, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Mikuláš Mlček
- Institute of Physiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Michaela Popková
- Institute of Physiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Tomáš Suchý
- Department of Composites and Carbon Materials, Institute of Rock Structure and Mechanics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; Department of Mechanics, Biomechanics and Mechatronics, Faculty of Mechanical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Monika Šupová
- Department of Composites and Carbon Materials, Institute of Rock Structure and Mechanics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jan Závora
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Václava Adámková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Hana Benáková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ondřej Slanař
- Department of Clinical Pharmacology and Pharmacy, Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Martin Šíma
- Department of Clinical Pharmacology and Pharmacy, Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Martin Bartoš
- Department of Stomatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Hynek Chlup
- Department of Mechanics, Biomechanics and Mechatronics, Faculty of Mechanical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Tomáš Grus
- 2nd Department of Cardiovascular Surgery, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
132
|
Yu Z, Smith MJ, Siow RCM, Liu KK. Ageing modulates human dermal fibroblast contractility: Quantification using nano-biomechanical testing. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118972. [PMID: 33515646 DOI: 10.1016/j.bbamcr.2021.118972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/19/2021] [Accepted: 01/24/2021] [Indexed: 12/12/2022]
Abstract
Dermal fibroblasts play a key role in maintaining homoeostasis and functionality of the skin. Their contractility plays a role in changes observed during ageing, especially in processes such as wound healing, inflammation, wrinkling and scar tissue formation as well as structural changes on extracellular matrix. Although alternations in skin physiology and morphology have been previously described, there remains a paucity of information about the influence of chronological ageing on dermal fibroblast contractility. In this study, we applied a novel nano-biomechanical technique on cell-embedded collagen hydrogels in combination with mathematical modelling and numerical simulation to measure contraction forces of normal human dermal fibroblasts (NHDF). We achieved quantitative differentiation of the contractility of cells derived from 'young' (< 30 years old) and 'aged' (> 60 years old) donors. Transforming growth factor β1 (TGF-β1) was used to stimulate the fibroblasts to assess their contractile potential. NHDF from aged donors exhibited a greater basal contractile force, while in contrast, NHDF from young donors have shown a significantly larger contractile force in response to TGF-β1 treatment. These findings validate our nano-biomechanical measurement technique and provide new insights for considering NHDF contractility in regenerative medicine and as a biomarker of dermal ageing processes.
Collapse
Affiliation(s)
- Zhuonan Yu
- School of Engineering, University of Warwick, Coventry, United Kingdom
| | - Matthew J Smith
- School of Cardiovascular Medicine & Sciences, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Richard C M Siow
- School of Cardiovascular Medicine & Sciences, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Kuo-Kang Liu
- School of Engineering, University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
133
|
Hennies HC, Poumay Y. Skin Disease Models In Vitro and Inflammatory Mechanisms: Predictability for Drug Development. Handb Exp Pharmacol 2021; 265:187-218. [PMID: 33387068 DOI: 10.1007/164_2020_428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Investigative skin biology, analysis of human skin diseases, and numerous clinical and pharmaceutical applications rely on skin models characterized by reproducibility and predictability. Traditionally, such models include animal models, mainly rodents, and cellular models. While animal models are highly useful in many studies, they are being replaced by human cellular models in more and more approaches amid recent technological development due to ethical considerations. The culture of keratinocytes and fibroblasts has been used in cell biology for many years. However, only the development of co-culture and three-dimensional epidermis and full-skin models have fundamentally contributed to our understanding of cell-cell interaction and cell signalling in the skin, keratinocyte adhesion and differentiation, and mechanisms of skin barrier function. The modelling of skin diseases has highlighted properties of the skin important for its integrity and cutaneous development. Examples of monogenic as well as complex diseases including atopic dermatitis and psoriasis have demonstrated the role of skin models to identify pathomechanisms and drug targets. Recent investigations have indicated that 3D skin models are well suitable for drug testing and preclinical studies of topical therapies. The analysis of skin diseases has recognized the importance of inflammatory mechanisms and immune responses and thus other cell types such as dendritic cells and T cells in the skin. Current developments include the production of more complete skin models comprising a range of different cell types. Organ models and even multi-organ systems are being developed for the analysis of higher levels of cellular interaction and drug responses and are among the most recent innovations in skin modelling. They promise improved robustness and flexibility and aim at a body-on-a-chip solution for comprehensive pharmaceutical in vitro studies.
Collapse
Affiliation(s)
- Hans Christian Hennies
- Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, UK. .,Cologne Center for Genomics, University Hospital Cologne, Cologne, Germany.
| | - Yves Poumay
- Faculty of Medicine, Namur Research Institute for Life Sciences, University of Namur, Namur, Belgium
| |
Collapse
|
134
|
Zhang Y, Ling Y, Zhang D, Wang M, Purslow C, Yang Y, Li C, Huang Z. Quantitative measurement of mechanical properties in wound healing processes in a corneal stroma model by using vibrational optical coherence elastography (OCE). BIOMEDICAL OPTICS EXPRESS 2021; 12:588-603. [PMID: 33659091 PMCID: PMC7899504 DOI: 10.1364/boe.404096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 05/11/2023]
Abstract
Corneal wound healing, caused by frequent traumatic injury to the cornea and increasing numbers of refractive surgeries, has become a vital clinical problem. In the cornea, wound healing is an extremely complicated process. However, little is known about how the biomechanical changes in wound healing response of the cornea. Collagen-based hydrogels incorporating corneal cells are suitable for replicating a three-dimensional (3D) equivalent of the cornea in-vitro. In this study, the mechanical properties of corneal stroma models were quantitatively monitored by a vibrational optical coherence elastography (OCE) system during continuous culture periods. Specifically, human corneal keratocytes were seeded at 5 × 105 cells/mL in the hydrogels with a collagen concentration of 3.0 mg/mL. The elastic modulus of the unwounded constructs increased from 2.950 ± 0.2 kPa to 11.0 ± 1.4 kPa, and the maximum thickness decreased from 1.034 ± 0.1 mm to 0.464 ± 0.09 mm during a 15-day culture period. Furthermore, a traumatic wound in the construct was introduced with a size of 500 µm. The elastic modulus of the neo-tissue in the wound area increased from 1.488 ± 0.4 kPa to 6.639 ± 0.3 kPa over 13 days. This study demonstrates that the vibrational OCE system is capable of quantitative monitoring the changes in mechanical properties of a corneal stroma wound model during continuous culture periods and improves our understanding on corneal wound healing processes.
Collapse
Affiliation(s)
- Yilong Zhang
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, Scotland, UK
| | - Yuting Ling
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, Scotland, UK
| | - Duo Zhang
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, Scotland, UK
| | - Mingkai Wang
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, Scotland, UK
| | - Christine Purslow
- Thea Pharmaceuticals Ltd, Keele University Science & Innovation Park, Innovation Way, Stoke-on-Trent, ST5 5NT, UK
| | - Ying Yang
- Guy Hilton Research Center, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Chunhui Li
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, Scotland, UK
| | - Zhihong Huang
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, Scotland, UK
| |
Collapse
|
135
|
Yang G, Liu H, Cui Y, Li J, Zhou X, Wang N, Wu F, Li Y, Liu Y, Jiang X, Zhang S. Bioinspired membrane provides periosteum-mimetic microenvironment for accelerating vascularized bone regeneration. Biomaterials 2020; 268:120561. [PMID: 33316630 DOI: 10.1016/j.biomaterials.2020.120561] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022]
Abstract
Periosteum plays a pivotal role in vascularization, ossification and remodeling during the healing process of bone injury. However, there are few studies focused on the construction of artificial implants with periosteum-mimetic effect. To emulate the primary role of natural periosteum or endosteal tissues in bone regeneration, here we provide a functional biomimetic membrane with micropatterns of site-specific biomineralization. The micropattern is generated by using printed hydroxyapatite nanoparticles (HANPs), combined with selective growth of biomineralized apatite and in situ coprecipitation with growth factors. The biomimetic membrane can sustainably provide a periosteum-mimetic microenvironment, such as long-term topographical guidance for cell recruitment and induced cell differentiation, by releasing calcium phosphate and growth factors. We demonstrated that rat mesenchymal stem cells (rMSCs) on such biomimetic membrane exhibited highly aligned organization, leading to enhanced angiogenesis and osteogenesis. In the rat calvarial defect model, our biomimetic membranes with biomineralized micropatterns could significantly enhance vascularized ossification and accelerate new bone formation. The current work suggests that the functionally biomimetic membranes with specific biomineralized micropatterns can be a promising alternative to periosteal autografts, with great potential for bench-to-bedside translation in orthopedics.
Collapse
Affiliation(s)
- Gaojie Yang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Media Lab and McGovern Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Haoming Liu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yi Cui
- Media Lab and McGovern Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jiaqi Li
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xuan Zhou
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Nuoxin Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Feige Wu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yan Li
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yu Liu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| | - Shengmin Zhang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
| |
Collapse
|
136
|
Elkhenany H, Elkodous MA, Newby SD, El-Derby AM, Dhar M, El-Badri N. Tissue Engineering Modalities and Nanotechnology. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/978-3-030-55359-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
137
|
Zhang SY, Hood M, Zhang IX, Chen CL, Zhang LL, Du J. Collagen and soy peptides attenuate contractile loss from UVA damage and enhance the antioxidant capacity of dermal fibroblasts. J Cosmet Dermatol 2020; 20:2277-2286. [PMID: 33095946 DOI: 10.1111/jocd.13805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Wrinkles and extracellular matrix (ECM) loss are common signs of skin aging and are thought to be the result of damage caused by reactive oxygen species (ROS); ROS induces an imbalance between ECM degradation and production. OBJECTIVES In this study, we evaluate soy peptides (SP) and collagen peptides (CP), alone and in combination, for their ability to inhibit ROS formation and increase ECM gene expression in order to ameliorate the signs of skin aging. METHODS Using tert-Butyl hydroperoxide (t-BuOOH)-treated dermal fibroblasts, we explored the potential of CP and SP to inhibit ROS formation by flow cytometry, as well as their effect on ECM component genes by real-time quantitative PCR. In addition, we examined the effect of CP and SP on UVA irradiated fibroblasts in a 3D collagen lattice model that measured contractility. RESULTS The results showed that the combination of CP and SP synergistically reduces ROS formation. This combination also increased expression of collagen I, collagen II, elastin, and fibronectin in t-BuOOH-treated or untreated dermal fibroblasts. In the UVA-treated 3D collagen lattice model, the results show that CP and SP significantly improved fibroblast contractility when compared to UVA control (P < 0.05). CONCLUSIONS In conclusion, CP and SP attenuate the loss of contractility due to UVA damage, inhibit t-BuOOH-induced ROS formation, and improve expression of ECM component genes.
Collapse
Affiliation(s)
- Sophia Y Zhang
- Amway (China) Innovation & Science Center, Shanghai, China
| | - Molly Hood
- Nutrilite Health Institute, Ada, MI, USA
| | - Iris X Zhang
- Amway (China) Innovation & Science Center, Shanghai, China
| | - Clark L Chen
- Amway (China) Innovation & Science Center, Shanghai, China
| | - Lynn L Zhang
- Amway (China) Innovation & Science Center, Shanghai, China
| | - Jun Du
- Amway (China) Innovation & Science Center, Shanghai, China
| |
Collapse
|
138
|
Stewart RH. A Modern View of the Interstitial Space in Health and Disease. Front Vet Sci 2020; 7:609583. [PMID: 33251275 PMCID: PMC7674635 DOI: 10.3389/fvets.2020.609583] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Increases in the volume of the interstitial space are readily recognized clinically as interstitial edema formation in the loose connective tissue of skin, mucosa, and lung. However, the contents and the hydrostatic pressure of this interstitial fluid can be very difficult to determine even in experimental settings. These difficulties have long obscured what we are beginning to appreciate is a dynamic milieu that is subject to both intrinsic and extrinsic regulation. This review examines current concepts regarding regulation of interstitial volume, pressure, and flow and utilizes that background to address three major topics of interest that impact IV fluid administration. The first of these started with the discovery that excess dietary salt can be stored non-osmotically in the interstitial space with minimal impact on vascular volume and pressures. This led to the hypothesis that, along with the kidney, the interstitial space plays an active role in the long-term regulation of blood pressure. Second, it now appears that hypovolemic shock leads to systemic inflammatory response syndrome principally through the entry of digestive enzymes into the intestinal interstitial space and the subsequent progression of enzymes and inflammatory agents through the mesenteric lymphatic system to the general circulation. Lastly, current evidence strongly supports the non-intuitive view that the primary factor leading to inflammatory edema formation is a decrease in interstitial hydrostatic pressure that dramatically increases microvascular filtration.
Collapse
Affiliation(s)
- Randolph H Stewart
- Department of Veterinary Physiology and Pharmacology, Michael E. DeBakey Institute, Texas A&M University, College Station, TX, United States
| |
Collapse
|
139
|
Tissue-scale tensional homeostasis in skin regulates structure and physiological function. Commun Biol 2020; 3:637. [PMID: 33127987 PMCID: PMC7603398 DOI: 10.1038/s42003-020-01365-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Tensional homeostasis is crucial for organ and tissue development, including the establishment of morphological and functional properties. Skin plays essential roles in waterproofing, cushioning and protecting deeper tissues by forming internal tension-distribution patterns, which involves aligning various cells, appendages and extracellular matrices (ECMs). The balance of traction force is thought to contribute to the formation of strong and pliable physical structures that maintain their integrity and flexibility. Here, by using a human skin equivalent (HSE), the horizontal tension-force balance of the dermal layer was found to clearly improve HSE characteristics, such as the physical relationship between cells and the ECM. The tension also promoted skin homeostasis through the activation of mechano-sensitive molecules such as ROCK and MRTF-A, and these results compared favourably to what was observed in tension-released models. Tension-induced HSE will contribute to analyze skin physiological functions regulated by tensional homeostasis as an alternative animal model.
Collapse
|
140
|
On collagen fiber morphoelasticity and homeostatic remodeling tone. J Mech Behav Biomed Mater 2020; 113:104154. [PMID: 33158790 DOI: 10.1016/j.jmbbm.2020.104154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 03/14/2020] [Accepted: 10/21/2020] [Indexed: 11/23/2022]
Abstract
A variety of biochemical and physical processes participate in the creation and maintenance of collagen in biological tissue. Under mechanical stimuli these collagen fibers undergo continuous processes of morphoelastic change. The model presented here is motivated by experimental reports of stretch-stabilization of the collagen fibers to enzymatic degradation. The fiber structure is modeled in terms of a fiber density evolution that is regulated by means of a fixed creation rate and a mechano-sensitive dissolution rate. The theory accounts for the possibly different natural configurations of the fiber unit constituents and the ground substance matrix. It also generalizes previous theoretical descriptions so as to account for finite survival times of the individual fiber units. Special consideration is given to steady state fiber-remodeling processes in which fiber creation and dissolution are in balance. Fiber assembly processes that involve prestretching the fiber constituents yield a homeostatic stress response with a characteristic fiber tone. Fiber density returns to homeostasis after mechanical disruption when sufficient time has passed.
Collapse
|
141
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
142
|
Yamanishi C, Parigoris E, Takayama S. Kinetic Analysis of Label-Free Microscale Collagen Gel Contraction Using Machine Learning-Aided Image Analysis. Front Bioeng Biotechnol 2020; 8:582602. [PMID: 33072731 PMCID: PMC7537788 DOI: 10.3389/fbioe.2020.582602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 09/02/2020] [Indexed: 11/17/2022] Open
Abstract
Pulmonary fibrosis is a deadly lung disease, wherein normal lung tissue is progressively replaced with fibrotic scar tissue. An aspect of this process can be recreated in vitro by embedding fibroblasts into a collagen matrix and providing a fibrotic stimulus. This work expands upon a previously described method to print microscale cell-laden collagen gels and combines it with live cell imaging and automated image analysis to enable high-throughput analysis of the kinetics of cell-mediated contraction of this collagen matrix. The image analysis method utilizes a plugin for FIJI, built around Waikato Environment for Knowledge Analysis (WEKA) Segmentation. After cross-validation of this automated image analysis with manual shape tracing, the assay was applied to primary human lung fibroblasts including cells isolated from idiopathic pulmonary fibrosis patients. In the absence of any exogenous stimuli, the analysis showed significantly faster and more extensive contraction of the diseased cells compared to the healthy ones. Upon stimulation with transforming growth factor beta 1 (TGF-β1), fibroblasts from the healthy donor showed significantly more contraction throughout the observation period while differences in the response of diseased cells was subtle and could only be detected during a smaller window of time. Finally, dose-response curves for the inhibition of collagen gel contraction were determined for 3 small molecules including the only 2 FDA-approved drugs for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Cameron Yamanishi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Eric Parigoris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
143
|
Tsukamoto Y, Akagi T, Akashi M. Supersensitive Layer-by-Layer 3D Cardiac Tissues Fabricated on a Collagen Culture Vessel Using Human-Induced Pluripotent Stem Cells. Tissue Eng Part C Methods 2020; 26:493-502. [PMID: 32873187 DOI: 10.1089/ten.tec.2020.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The fabrication of artificial cardiac tissue is an active area of research due to the shortage of donors for heart transplantation and for drug development. In our previous study, we fabricated vascularized three-dimensional (3D) cardiac tissue by layer-by-layer (LbL) and cell accumulation technique. However, it was not able to develop sufficient function because it was cultured on a hard plastic substrate. Experiment: Herein, we report the fabrication of high-performance 3D cardiac tissue by LbL and cell accumulation technique using a collagen culture vessel. Results: By using a collagen culture vessel, 3D cardiac tissue could be fabricated on a collagen culture vessel and this tissue showed high functionality due to improved interaction with the vessel. In the case of the plastic culture insert, 3D cardiac tissue was found to be peeled off, but this did not occur on the collagen culture vessel. In addition, the 3D cardiac tissue fabricated on a collagen culture vessel showed contraction that was 20 times larger than the tissue fabricated on a plastic culture insert. As a result of evaluation of cardiotoxicity using E-4031, the sensitivity of arrhythmia detection was increased by using collagen culture vessel. Conclusions: These results are expected to contribute to transplantation and drug discovery research as a 3D cardiac tissue model with a function similar to that of the living heart.
Collapse
Affiliation(s)
- Yoshinari Tsukamoto
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Takami Akagi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Mitsuru Akashi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
144
|
Wilcox DK, Bruner LH. In Vitro Alternatives for Ocular Safety Testing: An Outline of Assays and Possible Future Developments. Altern Lab Anim 2020. [DOI: 10.1177/026119299001800115.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
An increasing number of in vitro tests are being proposed as alternatives for animals in ocular safety testing. These in vitro systems use a variety of approaches and measure a broad range of endpoints, including cellular cytotoxicity and metabolism. We will briefly review the development and validation of these model systems. Several tests appear promising, and may prove useful as adjuncts in our current ocular safety assessment schemes. We will also discuss how these tests can most rapidly be brought to use in ocular safety assessment, and where efforts should be directed to develop more-highly predictive in vitro model systems for the future.
Collapse
Affiliation(s)
- David K. Wilcox
- Human & Environmental Safety Division, The Procter & Gamble Company, Miami Valley Laboratories, Cincinnati, OH 45239-8707, USA
| | - Leon H. Bruner
- Human & Environmental Safety Division, The Procter & Gamble Company, Miami Valley Laboratories, Cincinnati, OH 45239-8707, USA
| |
Collapse
|
145
|
Correa SO, Luo X, Raub CB. Microfluidic fabrication of stable collagen microgels with aligned microstructure using flow-driven co-deposition and ionic gelation. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2020; 30:085002. [PMID: 37273664 PMCID: PMC10237176 DOI: 10.1088/1361-6439/ab8ebf] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The controlled biofabrication of stable, aligned collagen hydrogels within microfluidic devices is critically important to the design of more physiologically accurate, longer-cultured on-chip models of tissue and organs. To address this goal, collagen-alginate microgels were formed in a microfluidic channel by calcium crosslinking of a flowing collagen-alginate solution through a cross-channel chitosan membrane spanning a pore allowing ion diffusion but not convection. The gels formed within seconds as isolated islands in a single channel, and their growth was self-limiting. Total gel thickness was controlled by altering the concentration of calcium and collagen-alginate flow rate to reach an equilibrium of calcium diffusion and solution convection at the gel boundary, for a desired thickness of 30-200 μm. Additionally, less calcium and higher flow produced greater compression of the gel, with regions farther from the pore compressing more. An aligned, stable collagen network was demonstrated by collagen birefringence, circumferential texture orientation, and little change in gel dimensions with de-chelation of calcium from alginate by prolonged flow of EDTA in the channel. Resultant gels were most stable and only slightly asymmetric when formed from solutions containing 8 mg ml-1 collagen. Diffusion of 4 kDa and 70 kDa fluorescently-labeled dextran indicated size-dependent diffusion across the gel, and accessibility of the construct to appropriately-sized bioactive molecules. This work demonstrates the physicochemical parameter control of collagen gel formation in microfluidic devices, with utility toward on-chip models of dense extracellular matrix invasion, cancer growth and drug delivery to cells within dense extracellular matrix bodies.
Collapse
Affiliation(s)
- Santiago O Correa
- Department of Biomedical Engineering, Washington DC, United States of America
| | - Xiaolong Luo
- Department of Mechanical Engineering, Washington DC, United States of America
- These authors contributed equally to this work
| | - Christopher B Raub
- Department of Biomedical Engineering, Washington DC, United States of America
- These authors contributed equally to this work
| |
Collapse
|
146
|
Derr K, Zou J, Luo K, Song MJ, Sittampalam GS, Zhou C, Michael S, Ferrer M, Derr P. Fully Three-Dimensional Bioprinted Skin Equivalent Constructs with Validated Morphology and Barrier Function. Tissue Eng Part C Methods 2020; 25:334-343. [PMID: 31007132 PMCID: PMC6589501 DOI: 10.1089/ten.tec.2018.0318] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Development of high-throughput, reproducible, three-dimensional (3D) bioprinted skin equivalents (BPSEs) that are morphologically and functionally comparable to native skin tissue is advancing research in skin diseases, and providing a physiologically relevant platform for the development of therapeutics, transplants for regenerative medicine, and testing of skin products like cosmetics. Current protocols for the production of engineered skin grafts are limited in their ability to control 3D geometry of the structure and contraction leading to variability of skin function between constructs. In this study, we describe a method for the biofabrication of skin equivalents (SEs) that are fully bioprinted using an open-market bioprinter, made with commercially available primary cells and natural hydrogels. The unique hydrogel formulation allows for the production of a human-like SE with minimal lateral tissue contraction in a multiwell plate format, thus making them suitable for high-throughput bioprinting in a single print with fast print and relatively short incubation times. The morphology and barrier function of the fully 3D BPSEs are validated by immunohistochemistry staining, optical coherence tomography, and permeation assays.
Collapse
Affiliation(s)
- Kristy Derr
- 1 Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Jinyun Zou
- 2 Department of Electrical and Computer Engineering, Bethlehem, Pennsylvania
| | - Keren Luo
- 2 Department of Electrical and Computer Engineering, Bethlehem, Pennsylvania
| | - Min Jae Song
- 3 National Eye Institute, National Institutes of Health, Rockville, Maryland
| | - G Sitta Sittampalam
- 1 Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Chao Zhou
- 2 Department of Electrical and Computer Engineering, Bethlehem, Pennsylvania.,4 Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania
| | - Sam Michael
- 1 Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Marc Ferrer
- 1 Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Paige Derr
- 1 Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
147
|
Gibb AA, Lazaropoulos MP, Elrod JW. Myofibroblasts and Fibrosis: Mitochondrial and Metabolic Control of Cellular Differentiation. Circ Res 2020; 127:427-447. [PMID: 32673537 DOI: 10.1161/circresaha.120.316958] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiac fibrosis is mediated by the activation of resident cardiac fibroblasts, which differentiate into myofibroblasts in response to injury or stress. Although myofibroblast formation is a physiological response to acute injury, such as myocardial infarction, myofibroblast persistence, as occurs in heart failure, contributes to maladaptive remodeling and progressive functional decline. Although traditional pathways of activation, such as TGFβ (transforming growth factor β) and AngII (angiotensin II), have been well characterized, less understood are the alterations in mitochondrial function and cellular metabolism that are necessary to initiate and sustain myofibroblast formation and function. In this review, we highlight recent reports detailing the mitochondrial and metabolic mechanisms that contribute to myofibroblast differentiation, persistence, and function with the hope of identifying novel therapeutic targets to treat, and potentially reverse, tissue organ fibrosis.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Michael P Lazaropoulos
- From the Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - John W Elrod
- From the Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
148
|
Shrestha S, McFadden MJ, Gramolini AO, Santerre JP. Proteome analysis of secretions from human monocyte-derived macrophages post-exposure to biomaterials and the effect of secretions on cardiac fibroblast fibrotic character. Acta Biomater 2020; 111:80-90. [PMID: 32428683 DOI: 10.1016/j.actbio.2020.04.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/31/2020] [Accepted: 04/23/2020] [Indexed: 12/27/2022]
Abstract
The use of exogenous biomolecules (BM) for the purpose of repairing and regenerating damaged cardiac tissue can yield serious side effects if used for prolonged periods. As well, such strategies can be cost prohibitive depending on the regiment and period of time applied. Alternatively, autologous monocytes/monocyte-derived macrophages (MDM) can provide a viable path towards generating an endogenous source of stimulatory BM. Biomaterials are often considered as delivery vehicles to generate unique profiles of such BM in tissues or to deliver autologous cells, that can influence the nature of BM produced by the cells. MDM cultured on a degradable polar hydrophobic ionic (D-PHI) polyurethane has previously demonstrated a propensity to increase select anti-inflammatory cytokines, and therefore there is good rationale to further investigate a broader spectrum of the cells' BM in order to provide a more complete proteomic analysis of human MDM secretions induced by D-PHI. Further, it is of interest to assess the potential of such BM to influence cells involved in the reparative state of vital tissues such as those that affect cardiac cell function. Hence, this current study examines the proteomic profile of MDM secretions using mass spectrometry for the first time, along with ELISA, following their culture on D-PHI, and compares them to two important reference materials, poly(lactic-co-glycolic acid) (PLGA) and tissue culture polystyrene (TCPS). Secretions collected from D-PHI cultured MDM led to higher levels of regenerative BM, AGRN, TGFBI and ANXA5, but lower levels of pro-fibrotic BM, MMP7, IL-1β, IL-6 and TNFα, when compared to MDM secretions collected from PLGA and TCPS. In the application to cardiac cell function, the secretion collected from D-PHI cultured MDM led to more human cardiac fibroblast (HCFs) migration. A lower collagen gel contraction induced by MDM secretions collected from D-PHI was supported by gene array analysis for human fibrosis-related genes. The implication of these findings is that more tailored biomaterials such as D-PHI, may lead to a lower pro-inflammatory phenotype of macrophages when used in cardiac tissue constructs, thereby enabling the development of vehicles for the delivery of interventional therapies, or be applied as coatings for sensor implants in cardiac tissue that minimize fibrosis. The general approach of using synthetic biomaterials in order to induce MDM secretions in a manner that will guide favorable regeneration will be critical in making the choice of biomaterials for tissue regeneration work in the future. STATEMENT OF SIGNIFICANCE: Immune modulation strategies currently applied in cardiac tissue repair are mainly based on the delivery of defined exogenous biomolecules. However, the use of such biomolecules may pose wide ranging systemic effects, thereby rendering them clinically less practical. The chemistry of biomaterials (used as a potential targeted delivery modality to circumvent the broad systemic effects of biomolecules) can not only affect acute and chronic toxicity but also alters the timeframe of the wound healing cascade. In this context, monocytes/monocyte-derive macrophages (MDM) can be harnessed as an immune modulating strategy to promote wound healing by an appropriate choice of the biomaterial. However, there are limited reports on the complete proteome analysis of MDM and their reaction of biomaterial related interventions on cardiac tissues and cells. No studies to date have demonstrated the complete proteome of MDM secretions when these cells were cultured on a non-traditional immune modulatory ionomeric polyurethane D-PHI film. This study demonstrated that MDM cultured on D-PHI expressed significantly higher levels of AGRN, TGFBI and ANXA5 but lower levels of MMP7, IL-1β, IL-6 and TNFα when compared to MDM cultured on a well-established degradable biomaterials in the medical field, e.g. PLGA and TCPS, which are often used as the relative standards for cell culture work in the biomaterials field. The implications of these findings have relevance to the repair of cardiac tissues. In another aspect of the work, human cardiac fibroblasts showed significantly lower contractility (low collagen gel contraction and low levels of ACTA2) when cultured in the presence of MDM secretions collected after culturing them on D-PHI compared to PLGA and TCPS. The findings place emphasis on the importance of making the choice of biomaterials for tissue engineering and regenerative medicine applied to their use in cardiac tissue repair.
Collapse
Affiliation(s)
- Suja Shrestha
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada
| | - Meghan J McFadden
- Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1M8, Canada
| | - J Paul Santerre
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program and Ted Rogers Centre for Heart Research, Toronto, Ontario M5G 1M1, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada.
| |
Collapse
|
149
|
Montero A, Acosta S, Hernández R, Elvira C, Jorcano JL, Velasco D. Contraction of fibrin-derived matrices and its implications for in vitro human skin bioengineering. J Biomed Mater Res A 2020; 109:500-514. [PMID: 32506782 DOI: 10.1002/jbm.a.37033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/09/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
It is well-known that fibroblasts play a fundamental role in the contraction of collagen and fibrin hydrogels when used in the production of in vitro bilayered skin substitutes. However, little is known about the contribution of other factors, such as the hydrogel matrix itself, on this contraction. In this work, we studied the contraction of plasma-derived fibrin hydrogels at different temperatures (4, 23, and 37°C) in an isotonic buffer (phosphate-buffered saline). These types of hydrogels presented a contraction of approximately 30% during the first 24 hr, following a similar kinetics irrespectively of the temperature. This kinetics continued in a slowed down manner to reach a plateau value of 40% contraction after 10-15 days. Contraction of commercial fibrinogen hydrogels was studied under similar conditions and the kinetics was completed after 8 hr, reaching values between 20 and 70% depending on the temperature. We attribute these substantial differences to a modulatory effect on the contraction due to plasma proteins which are initially embedded in, and progressively released from, the plasma-based hydrogels. The elastic modulus of hydrogels measured at a constant frequency decreased with increasing temperature in 7-day gels. Rheological measurements showed the absence of a strain-hardening behavior in the plasma-derived fibrin hydrogels. Finally, plasma-derived fibrin hydrogels with and without human primary fibroblast and keratinocytes were prepared in transwell inserts and their height measured over time. Both cellular and acellular gels showed a height reduction of 30% during the first 24 hr likely due to the above-mentioned intrinsic fibrin matrix contraction.
Collapse
Affiliation(s)
- Andrés Montero
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
| | - Sonia Acosta
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
| | - Rebeca Hernández
- Institute of Polymer Science and Technology, CSIC, Madrid, Spain
| | - Carlos Elvira
- Department of Applied Macromolecular Chemistry, Institute of Polymer Science and Technology, CSIC, Madrid, Spain
| | - José Luis Jorcano
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), Madrid, Spain.,Division of Epithelial Biomedicine, CIEMAT, Madrid, Spain
| | - Diego Velasco
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
| |
Collapse
|
150
|
Cambria E, Brunner S, Heusser S, Fisch P, Hitzl W, Ferguson SJ, Wuertz-Kozak K. Cell-Laden Agarose-Collagen Composite Hydrogels for Mechanotransduction Studies. Front Bioeng Biotechnol 2020; 8:346. [PMID: 32373605 PMCID: PMC7186378 DOI: 10.3389/fbioe.2020.00346] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
The increasing investigation of cellular mechanotransduction mechanisms requires biomaterials combining biofunctionality and suitable mechanical properties. Agarose is a standard biomaterial for cartilage and intervertebral disc mechanobiology studies, but lacks adhesion motifs and the necessary cell-matrix interaction for mechanotransduction. Here, collagen type I was blended at two concentrations (2 and 4.5 mg/mL) with agarose 2% wt/vol. The composite hydrogels were characterized in terms of structural homogeneity, rheological properties and size stability. Nucleus pulposus (NP) cell viability, proliferation, morphology, gene expression, GAG production, adhesion and mechanotransduction ability were further tested. Blended hydrogels presented a homogenous network of the two polymers. While the addition of 4.5 mg/mL collagen significantly decreased the storage modulus and increased the loss modulus of the gels, blended gels containing 2 mg/mL collagen displayed similar mechanical properties to agarose. Hydrogel size was conserved over 21 days for all agarose-based gels. Embedded cells were viable (>80%) and presented reduced proliferation and a round morphology typical of NP cells in vivo. Gene expression of collagen types I and II and aggrecan significantly increased in blended hydrogels from day 1 to 7, further resulting in a significantly superior GAG/DNA ratio compared to agarose gels at day 7. Agarose-collagen hydrogels not only promoted cell adhesion, contrary to agarose gels, but also showed a 5.36-fold higher focal adhesion kinase phosphorylation (pFAK/β-tubulin) when not compressed, and increased pFAK/FAK values 10 min after compression. Agarose-collagen thus outperforms agarose, mimics native tissues constituted of non-fibrillar matrix and collagens, and allows exploring complex loading in a highly reproducible system.
Collapse
Affiliation(s)
- Elena Cambria
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Silvio Brunner
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Sally Heusser
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Philipp Fisch
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Wolfgang Hitzl
- Research Office (Biostatistics), Paracelsus Medical University, Salzburg, Austria.,Department of Ophthalmology and Optometry, Paracelsus Medical University, Salzburg, Austria.,Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | | | - Karin Wuertz-Kozak
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.,Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States.,Spine Center, Schön Klinik München Harlaching, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Private Medical University Salzburg (Austria), Munich, Germany
| |
Collapse
|