1
|
Cariba S, Srivastava A, Bronsema K, Kouthouridis S, Zhang B, Payne SL. Innervated Coculture Device to Model Peripheral Nerve-Mediated Fibroblast Activation. ACS Biomater Sci Eng 2024; 10:7566-7576. [PMID: 39601321 PMCID: PMC11633653 DOI: 10.1021/acsbiomaterials.4c01482] [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: 08/13/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Cutaneous wound healing is a complex process involving various cellular and molecular interactions, resulting in the formation of a collagen-rich scar with imperfect function and morphology. Dermal fibroblasts are crucial to successful wound healing, migrating to the wound site where they are activated to provide extracellular matrix remodeling and wound closure. Peripheral nerves have been shown to play an important role in wound healing, with loss or damage to these nerves often leading to impaired healing and the formation of chronic nonhealing wounds. Previous research has suggested that sensory nerves secrete trophic factors that can regulate wound healing, including fibroblast activation; however, the direct cell-cell interaction between nerves and fibroblasts has not been extensively studied. To address this knowledge gap, we developed an in vitro co-culture model using a device called the IFlowPlate. This model supports the long-term viability of multiple cell types while allowing for direct contact between sensory nerve cells and dermal fibroblasts. Using the IFlowPlate, we demonstrate that co-culture of dorsal root ganglia with dermal fibroblasts increases fibroblast proliferation, collagen and α-smooth muscle actin expression, and secretion of pro-wound healing factors, suggesting that nerves can promote wound healing by modulating fibroblast activation. The IFlowPlate offers a user-friendly and high-throughput platform to study the in vitro interactions between nerves and a variety of cell types that can be applied to wound healing and other important biological processes.
Collapse
Affiliation(s)
- Solsa Cariba
- Department
of Biomedical Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Avika Srivastava
- Department
of Biomedical Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Kendra Bronsema
- Department
of Biomedical Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Sonya Kouthouridis
- Department
of Chemical Engineering, McMaster University, Hamilton L8S 4L8, Canada
| | - Boyang Zhang
- Department
of Chemical Engineering, McMaster University, Hamilton L8S 4L8, Canada
- School
of Biomedical Engineering, McMaster University, Hamilton L8S 4L8, Canada
| | - Samantha L. Payne
- Department
of Biomedical Sciences, University of Guelph, Guelph N1G 2W1, Canada
| |
Collapse
|
2
|
Fois MG, Tahmasebi Birgani ZN, López-Iglesias C, Knoops K, van Blitterswijk C, Giselbrecht S, Habibović P, Truckenmüller RK. In vitro vascularization of 3D cell aggregates in microwells with integrated vascular beds. Mater Today Bio 2024; 29:101260. [PMID: 39391792 PMCID: PMC11466645 DOI: 10.1016/j.mtbio.2024.101260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/20/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Most human tissues possess vascular networks supplying oxygen and nutrients. Engineering of functional tissue and organ models or equivalents often require the integration of artificial vascular networks. Several approaches, such as organs on chips and three-dimensional (3D) bioprinting, have been pursued to obtain vasculature and vascularized tissues in vitro. This technical feasibility study proposes a new approach for the in vitro vascularization of 3D microtissues. For this, we thermoform arrays of round-bottom microwells into thin non-porous and porous polymer films/membranes and culture vascular beds on them from which endothelial sprouting occurs in a Matrigel-based 3D extra cellular matrix. We present two possible culture configurations for the microwell-integrated vascular beds. In the first configuration, human umbilical vein endothelial cells (HUVECs) grow on and sprout from the inner wall of the non-porous microwells. In the second one, HUVECs grow on the outer surface of the porous microwells and sprout through the pores toward the inside. These approaches are extended to lymphatic endothelial cells. As a proof of concept, we demonstrate the in vitro vascularization of spheroids from human mesenchymal stem cells and MG-63 human osteosarcoma cells. Our results show the potential of this approach to provide the spheroids with an abundant outer vascular network and the indication of an inner vasculature.
Collapse
Affiliation(s)
- Maria G. Fois
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Zeinab N. Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
3
|
Simonyan H, Palumbo R, Petrosyan S, Mkrtchyan A, Galstyan A, Saghyan A, Scognamiglio PL, Vicidomini C, Fik-Jaskólka M, Roviello GN. BSA Binding and Aggregate Formation of a Synthetic Amino Acid with Potential for Promoting Fibroblast Proliferation: An In Silico, CD Spectroscopic, DLS, and Cellular Study. Biomolecules 2024; 14:579. [PMID: 38785986 PMCID: PMC11118884 DOI: 10.3390/biom14050579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
This study presents the chemical synthesis, purification, and characterization of a novel non-natural synthetic amino acid. The compound was synthesized in solution, purified, and characterized using NMR spectroscopy, polarimetry, and melting point determination. Dynamic Light Scattering (DLS) analysis demonstrated its ability to form aggregates with an average size of 391 nm, extending to the low micrometric size range. Furthermore, cellular biological assays revealed its ability to enhance fibroblast cell growth, highlighting its potential for tissue regenerative applications. Circular dichroism (CD) spectroscopy showed the ability of the synthetic amino acid to bind serum albumins (using bovine serum albumin (BSA) as a model), and CD deconvolution provided insights into the changes in the secondary structures of BSA upon interaction with the amino acid ligand. Additionally, molecular docking using HDOCK software elucidated the most likely binding mode of the ligand inside the BSA structure. We also performed in silico oligomerization of the synthetic compound in order to obtain a model of aggregate to investigate computationally. In more detail, the dimer formation achieved by molecular self-docking showed two distinct poses, corresponding to the lowest and comparable energies, with one pose exhibiting a quasi-coplanar arrangement characterized by a close alignment of two aromatic rings from the synthetic amino acids within the dimer, suggesting the presence of π-π stacking interactions. In contrast, the second pose displayed a non-coplanar configuration, with the aromatic rings oriented in a staggered arrangement, indicating distinct modes of interaction. Both poses were further utilized in the self-docking procedure. Notably, iterative molecular docking of amino acid structures resulted in the formation of higher-order aggregates, with a model of a 512-mer aggregate obtained through self-docking procedures. This model of aggregate presented a cavity capable of hosting therapeutic cargoes and biomolecules, rendering it a potential scaffold for cell adhesion and growth in tissue regenerative applications. Overall, our findings highlight the potential of this synthetic amino acid for tissue regenerative therapeutics and provide valuable insights into its molecular interactions and aggregation behavior.
Collapse
Affiliation(s)
- Hayarpi Simonyan
- Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., Yerevan 0025, Armenia
| | - Rosanna Palumbo
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Satenik Petrosyan
- Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., Yerevan 0025, Armenia
| | - Anna Mkrtchyan
- Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., Yerevan 0025, Armenia
| | - Armen Galstyan
- Department of Chemistry, Yerevan State University, 1 Alex Manoogian Str., Yerevan 0025, Armenia
| | - Ashot Saghyan
- Institute of Pharmacy, Yerevan State University, 1 Alex Manoogian Str., Yerevan 0025, Armenia
| | | | - Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Marta Fik-Jaskólka
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Council for Research (IBB-CNR), Area di Ricerca Site and Headquarters, Via Pietro Castellino 111, 80131 Naples, Italy
| |
Collapse
|
4
|
Kitana W, Levario‐Diaz V, Cavalcanti‐Adam EA, Ionov L. Biofabrication of Composite Bioink-Nanofiber Constructs: Effect of Rheological Properties of Bioinks on 3D (Bio)Printing and Cells Interaction with Aligned Touch Spun Nanofibers. Adv Healthc Mater 2024; 13:e2303343. [PMID: 38009530 PMCID: PMC11469018 DOI: 10.1002/adhm.202303343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Indexed: 11/29/2023]
Abstract
This paper reports on a novel approach for the fabrication of composite multilayered bioink-nanofibers construct. This work achieves this by using a hands-free 3D (bio)printing integrated touch-spinning approach. Additionally, this work investigates the interaction of fibroblasts in different bioinks with the highly aligned touch-spun nanofibers. This work conducts a comprehensive characterization of the rheological properties of the inks, starting with low-strain oscillatory rheology to analyze the viscoelastic behavior, when the material structure remains intact. Moreover, this work performs amplitude sweeps to investigate the stability of the inks under large deformations, rotational rheology to examine the shear thinning profile, and a three-step creep experiment to study time-dependent rheological behavior. The obtained rheological results are correlated to visual observation of the flow behavior of inks. These behaviors span from an ink with zero-shear viscosity, very weak shear thinning, and no thixotropic behavior to inks exhibiting flow stress, pronounced shear thinning, and thixotropy. It is demonstrated that inks have an essential effect on cell behavior. While all bioinks allow a preferred directionality of the fibroblasts along the fiber direction, cells tend to form aggregates in bioinks with higher viscosity, and a considerable number of agglomerates are observed in the presence of laponite-RD.
Collapse
Affiliation(s)
- Waseem Kitana
- Professorship of BiofabricationFaculty of Engineering ScienceUniversity of BayreuthLudwig‐Thoma‐Straße 36A95447BayreuthGermany
| | - Victoria Levario‐Diaz
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 2969120HeidelbergGermany
| | - Elisabetta Ada Cavalcanti‐Adam
- Department of Cellular BiophysicsMax Planck Institute for Medical ResearchJahnstraße 2969120HeidelbergGermany
- Professorship of Cellular BiomechanicsFaculty of Engineering ScienceUniversity of BayreuthUniversitätsstraße 3095447BayreuthGermany
| | - Leonid Ionov
- Professorship of BiofabricationFaculty of Engineering ScienceUniversity of BayreuthLudwig‐Thoma‐Straße 36A95447BayreuthGermany
- Bavarian Polymer InstituteUniversity of BayreuthUniversitätsstraße 3095447BayreuthGermany
| |
Collapse
|
5
|
Pien N, Di Francesco D, Copes F, Bartolf-Kopp M, Chausse V, Meeremans M, Pegueroles M, Jüngst T, De Schauwer C, Boccafoschi F, Dubruel P, Van Vlierberghe S, Mantovani D. Polymeric reinforcements for cellularized collagen-based vascular wall models: influence of the scaffold architecture on the mechanical and biological properties. Front Bioeng Biotechnol 2023; 11:1285565. [PMID: 38053846 PMCID: PMC10694796 DOI: 10.3389/fbioe.2023.1285565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
A previously developed cellularized collagen-based vascular wall model showed promising results in mimicking the biological properties of a native vessel but lacked appropriate mechanical properties. In this work, we aim to improve this collagen-based model by reinforcing it using a tubular polymeric (reinforcement) scaffold. The polymeric reinforcements were fabricated exploiting commercial poly (ε-caprolactone) (PCL), a polymer already used to fabricate other FDA-approved and commercially available devices serving medical applications, through 1) solution electrospinning (SES), 2) 3D printing (3DP) and 3) melt electrowriting (MEW). The non-reinforced cellularized collagen-based model was used as a reference (COL). The effect of the scaffold's architecture on the resulting mechanical and biological properties of the reinforced collagen-based model were evaluated. SEM imaging showed the differences in scaffolds' architecture (fiber alignment, fiber diameter and pore size) at both the micro- and the macrolevel. The polymeric scaffold led to significantly improved mechanical properties for the reinforced collagen-based model (initial elastic moduli of 382.05 ± 132.01 kPa, 100.59 ± 31.15 kPa and 245.78 ± 33.54 kPa, respectively for SES, 3DP and MEW at day 7 of maturation) compared to the non-reinforced collagen-based model (16.63 ± 5.69 kPa). Moreover, on day 7, the developed collagen gels showed stresses (for strains between 20% and 55%) in the range of [5-15] kPa for COL, [80-350] kPa for SES, [20-70] kPa for 3DP and [100-190] kPa for MEW. In addition to the effect on the resulting mechanical properties, the polymeric tubes' architecture influenced cell behavior, in terms of proliferation and attachment, along with collagen gel compaction and extracellular matrix protein expression. The MEW reinforcement resulted in a collagen gel compaction similar to the COL reference, whereas 3DP and SES led to thinner and longer collagen gels. Overall, it can be concluded that 1) the selected processing technique influences the scaffolds' architecture, which in turn influences the resulting mechanical and biological properties, and 2) the incorporation of a polymeric reinforcement leads to mechanical properties closely matching those of native arteries.
Collapse
Affiliation(s)
- Nele Pien
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC, Canada
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Dalila Di Francesco
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC, Canada
- Laboratory of Human Anatomy, Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Francesco Copes
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC, Canada
| | - Michael Bartolf-Kopp
- Department of Functional Materials in Medicine and Dentistry, Institute of Biofabrication and Functional Materials, University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg, Germany
| | - Victor Chausse
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Marguerite Meeremans
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Marta Pegueroles
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Tomasz Jüngst
- Department of Functional Materials in Medicine and Dentistry, Institute of Biofabrication and Functional Materials, University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg, Germany
| | - Catharina De Schauwer
- Faculty of Veterinary Medicine, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Francesca Boccafoschi
- Laboratory of Human Anatomy, Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, QC, Canada
| |
Collapse
|
6
|
Anitua E, Zalduendo M, Troya M, Tierno R, Alkhraisat MH. Cellular composition modifies the biological properties and stability of platelet rich plasma membranes for tissue engineering. J Biomed Mater Res A 2023; 111:1710-1721. [PMID: 37318048 DOI: 10.1002/jbm.a.37579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 03/03/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
Scaffolds should provide structural support for tissue regeneration, allowing their gradual biodegradation and interacting with cells and bioactive molecules to promote remodeling. Thus, the scaffold's intrinsic properties affect cellular processes involved in tissue regeneration, including migration, proliferation, differentiation, and protein synthesis. In this sense, due to its biological effect and clinical potential, Platelet Rich Plasma (PRP) fibrin could be considered a successful scaffold. Given the high variability in commercial PRPs formulations, this research focused on assessing the influence of cellular composition on fibrin membrane stability and remodeling cell activity. The stability and biological effect were evaluated at different time points via D-dimer, type I collagen and elastase quantification in culture media conditioned by Plasma Rich in Growth Factors - Fraction 1 (PRGF-F1), Plasma Rich in Growth Factors - Whole Plasma (PRGF-WP) and Leukocyte-rich Platelet Rich Plasma (L-PRP) membranes, and by gingival fibroblast cells seeded on them, respectively. Ultrastructure of PRP membranes was also evaluated. Histological analyses were performed after 5 and 18 days. Additionally, the effect of fibrin membranes on cell proliferation was determined. According to the results, L-PRP fibrin membranes degradation was complete at the end of the study, while PRGF membranes remained practically unchanged. Considering fibroblast behavior, PRGF membranes, in contrast to L-PRP ones, promoted extracellular matrix biosynthesis at the same time as fibrinolysis and enhanced cell proliferation. In conclusion, leukocytes in PRP fibrin membranes drastically reduce scaffold stability and induce behavioral changes in fibroblasts by reducing their proliferation rate and remodeling ability.
Collapse
Affiliation(s)
- Eduardo Anitua
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | - Mar Zalduendo
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | - María Troya
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | - Roberto Tierno
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | | |
Collapse
|
7
|
Binlateh T, Hutamekalin P, Yongsawatdigul J, Yamabhai M, Jitprasertwong P. Effects of collagen, chitosan and mixture on fibroblast responses and angiogenic activities in 2D and 3D in vitro models. J Biomed Mater Res A 2023; 111:1642-1655. [PMID: 37222462 DOI: 10.1002/jbm.a.37561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/21/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023]
Abstract
Despite accumulating evidences have demonstrated the potential of collagen and chitosan on tissue repair, it remains unclear on their combination effects. Here, we examined the regenerative effects of single collagen, chitosan and their mixture on fibroblasts and endothelial cells at cellular levels. The results showed that fibroblast responses, as indicated by high proliferative rate, increased spheroid diameter and migrated area existing from spheroid edge, and decreased wound area, were significantly promoted by either collagen or chitosan stimulation. Similarly, both collagen and chitosan resulted in increased endothelial cell proliferation and migration with accelerated tube-like network formation and upregulated VE-cadherin expression, although collagen strongly provided this effect. While the 1:1 mixture (100:100 μg/mL of chitosan to collagen) treatment caused a reduction in fibroblast viability, the lower ratio of chitosan (1:10 mixture; 10:100 μg/mL) did not produce any impact on both fibroblast and endothelial cell viabilities. The 1:10 mixture also significantly enhanced the additional effects on fibroblast responses and angiogenic activities as shown by higher endothelial growth, proliferation and migration with accelerated capillary-like network formation than those treated with the single substance. Further investigation of signaling proteins found that collagen significantly increased expressions of p-Fak, p-Akt and Cdk5 whereas chitosan upregulated p-Fak and Cdk5 expressions. Comparing to the single treatments, p-Fak, p-Akt and Cdk5 were higher expressed in the 1:10 mixture. These observations indicate that proper collagen-chitosan mixture provides the combination effects on fibroblast responses and angiogenic activities when a high concentration of collagen is used, possibly through Fak/Akt and Cdk5 signaling pathways. Therefore, this study helps to define the clinical use of collagen and chitosan as promising biomaterials for tissue repair.
Collapse
Affiliation(s)
- Thunwa Binlateh
- School of Pharmacy, Walailak University, Nakhon Si Thammarat, Thailand
| | - Pilaiwanwadee Hutamekalin
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Jirawat Yongsawatdigul
- Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Montarop Yamabhai
- Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | | |
Collapse
|
8
|
Ryan CN, Pugliese E, Shologu N, Gaspar D, Rooney P, Islam MN, O'Riordan A, Biggs MJ, Griffin MD, Zeugolis DI. Physicochemical cues are not potent regulators of human dermal fibroblast trans-differentiation. BIOMATERIALS AND BIOSYSTEMS 2023; 11:100079. [PMID: 37720487 PMCID: PMC10499661 DOI: 10.1016/j.bbiosy.2023.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/25/2023] [Accepted: 05/29/2023] [Indexed: 09/19/2023] Open
Abstract
Due to their inherent plasticity, dermal fibroblasts hold great promise in regenerative medicine. Although biological signals have been well-established as potent regulators of dermal fibroblast function, it is still unclear whether physiochemical cues can induce dermal fibroblast trans-differentiation. Herein, we evaluated the combined effect of surface topography, substrate rigidity, collagen type I coating and macromolecular crowding in human dermal fibroblast cultures. Our data indicate that tissue culture plastic and collagen type I coating increased cell proliferation and metabolic activity. None of the assessed in vitro microenvironment modulators affected cell viability. Anisotropic surface topography induced bidirectional cell morphology, especially on more rigid (1,000 kPa and 130 kPa) substrates. Macromolecular crowding increased various collagen types, but not fibronectin, deposition. Macromolecular crowding induced globular extracellular matrix deposition, independently of the properties of the substrate. At day 14 (longest time point assessed), macromolecular crowding downregulated tenascin C (in 9 out of the 14 groups), aggrecan (in 13 out of the 14 groups), osteonectin (in 13 out of the 14 groups), and collagen type I (in all groups). Overall, our data suggest that physicochemical cues (such surface topography, substrate rigidity, collagen coating and macromolecular crowding) are not as potent as biological signals in inducing dermal fibroblast trans-differentiation.
Collapse
Affiliation(s)
- Christina N.M. Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Naledi Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Peadar Rooney
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Md Nahidul Islam
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, University of Galway, Galway, Ireland
- Discipline of Biochemistry, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Alan O'Riordan
- Tyndall National Institute, University College Cork (UCC), Cork, Ireland
| | - Manus J. Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
9
|
Rivas J, Dubois A, Blanquer A, Gérardy M, Ziegler U, Groschup MH, Grobet L, Garigliany MM. Tendon-Derived Mesenchymal Stem Cells (TDSCs) as an In Vitro Model for Virological Studies in Wild Birds. Viruses 2023; 15:1455. [PMID: 37515142 PMCID: PMC10383174 DOI: 10.3390/v15071455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
The use of wild animals in research is complicated due to the capture and housing conditions, as well as to legal aspects, making it difficult to develop in vivo and in vitro models for the study of pathologies that affect these species. Here we validate an in vitro model of tendon-derived mesenchymal cells (TDSC) from Eurasian blackbird (Turdus merula) cadaveric samples. Through the expression of surface markers and the ability to differentiate into multiple lineages, the nature of the cells was confirmed. We then evaluated Mesenchymal Stem Cells (MSCs) as an infection model for the Usutu Flavivirus. To this aim, blackbird TDSCs were compared to Vero E6 cells, commonly used in Flavivirus studies. Both cells showed permissiveness to USUV infection as confirmed by immunocytochemistry. Moreover, TDSCs exhibited replication kinetics similar to, although slightly lower than, Vero E6, confirming these cells as a pertinent study model for the study of the pathogenesis of USUV. In this work, we isolated and characterized tendon-derived mesenchymal stem cells, which represent an interesting and convenient in vitro model for the study of wildlife species in laboratories.
Collapse
Affiliation(s)
- José Rivas
- Fundamental and Applied Research for Animals & Health (FARAH), Laboratory of Pathology, Faculty of Veterinary Medicine, University of Liège, Sart Tilman B43, B-4000 Liège, Belgium; (J.R.); (A.B.); (M.G.)
| | - Axel Dubois
- Fundamental and Applied Research for Animals & Health (FARAH), Laboratory of Embryology, Faculty of Veterinary Medicine, University of Liège, Sart Tilman B43, B-4000 Liège, Belgium; (A.D.); (L.G.)
| | - Aude Blanquer
- Fundamental and Applied Research for Animals & Health (FARAH), Laboratory of Pathology, Faculty of Veterinary Medicine, University of Liège, Sart Tilman B43, B-4000 Liège, Belgium; (J.R.); (A.B.); (M.G.)
| | - Mazarine Gérardy
- Fundamental and Applied Research for Animals & Health (FARAH), Laboratory of Pathology, Faculty of Veterinary Medicine, University of Liège, Sart Tilman B43, B-4000 Liège, Belgium; (J.R.); (A.B.); (M.G.)
| | - Ute Ziegler
- Institute for Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (U.Z.); (M.H.G.)
| | - Martin H. Groschup
- Institute for Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (U.Z.); (M.H.G.)
| | - Luc Grobet
- Fundamental and Applied Research for Animals & Health (FARAH), Laboratory of Embryology, Faculty of Veterinary Medicine, University of Liège, Sart Tilman B43, B-4000 Liège, Belgium; (A.D.); (L.G.)
| | - Mutien-Marie Garigliany
- Fundamental and Applied Research for Animals & Health (FARAH), Laboratory of Pathology, Faculty of Veterinary Medicine, University of Liège, Sart Tilman B43, B-4000 Liège, Belgium; (J.R.); (A.B.); (M.G.)
| |
Collapse
|
10
|
Rachinskaya OA, Melnikova EV, Merkulov VA. FEATURES OF QUALITY CONTROL STRATEGY FOR DRUGS BASED ON VIABLE SKIN CELLS. PHARMACY & PHARMACOLOGY 2023. [DOI: 10.19163/2307-9266-2022-10-6-515-524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The aim of the study was to research the international experience in quality assurance of the products based on skin cells in order to identify the features of the quality control strategy in the development, production, as well as during an expert quality assessment as a part of the state registration procedure in the Russian Federation.Materials and methods. The article provides an analysis of the materials presented in the assessment reports of the USA and Japanese regulatory authorities, as well as on the official websites of manufacturers, in review and scientific papers on the study of the structure and properties of tissue-engineered skin analogs.Results. The manufacture of products containing human skin cells is associated with such risks as the possibility of contamination of the preparation with infective agents transmitted by materials of the animal origin, feeder cells, donor cells, or during the manufacturing process; a small amount of biopsy materials; a complexity of a three-dimensional product structure when combining cells with a scaffold; continuity of the manufacture process and a short product expiry date. The raw materials and reagents control, the creation of cell banks, using animal feeder cells only from qualified cell banks, an in-process control and release testing in accordance with the requirements of the finished product specification, make it possible to obtain a preparation with a reproducible quality. The specification should contain information about the identity, safety and potency of the product. For each preparation, the choice of approaches for assessing the quality is individual and depends on its composition and mode of action.Conclusion. The features of the quality control strategy for the drugs based on human skin cells, consist in the implementation of control measures in order to obtain a proper quality of cellular (viability, sterility, identity, potency, et al) and non-cellular (physico-chemical scaffold properties) components or the whole graft (bioburden, barrier properties). The approaches and methods for determining the potency should be selected individually for each product and reflect the number, viability and identity of cells, a proliferative activity and secretable ability of the cellular component.
Collapse
Affiliation(s)
| | - E. V. Melnikova
- Scientific Centre for Expert Evaluation of Medicinal Products
| | - V. A. Merkulov
- Scientific Centre for Expert Evaluation of Medicinal Products
| |
Collapse
|
11
|
Silk Fibroin Hybrids for Biological Scaffolds with Adhesive Surface and Adaptability to the Target Tissue Change. THE EUROBIOTECH JOURNAL 2023. [DOI: 10.2478/ebtj-2023-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
Background Regenerative Medicine (RM) is a branch of medicine that aims to regenerate tissues and organs to overcome the problems transplants entail (poor availability, risk of rejection and intense immunosuppression). To do this, RM makes use of tissue engineering (TE). This fundamental branch deals with creating biological scaffolds capable of performing the role that physiologically belongs to the extracellular matrix (ECM). In this review, we report how specific characteristics of the scaffolds (bio-compatibility, biodegradability and mechanical and conformal properties) can be obtained using 3D printing, which facilitates the emulation of physiological tissues and organs.
Purpose and scope This review reports recent advances in the fabrication method of bioactive scaffolds that can be used clinically, providing support for cell seeding and proliferation. To this end, silk fibroin, tannin and graphene were used to improve the scaffold’s electro-bio-mechanical properties. These materials in different compositions are studied to demonstrate their potential use as bio-ink in bioadhesives and cellularized and implantable 3D-printed scaffolds.
Summary of new synthesis and conclusions reached in the review Silk fibroin is a natural biopolymer; tannin, on the other hand, is a biological polyphenol, highly reactive with other molecules by nature and with promising antioxidant capabilities. Finally, graphene is nothing more than a monolayer of graphite that has been shown to implement the mechanics and electrical conductivity of the compounds in which it is inserted; it also has excellent biocompatibility and surface area, qualities that promote cell adhesion and growth.
Conclusion Polyphenols and graphene have been shown to work in synergy in improving the electro-mechanical properties of silk fibroin scaffolds. We reported optimal and potentially market-competitive bioadhesives, but above all, the proliferation of neuronal precursor cells in vitro was successfully demonstrated.
Collapse
|
12
|
[Effect of fibroblasts on promoting the sprout and migration of endothelial cells in three-dimensional pre-vascularized microstructures]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:881-888. [PMID: 35848186 PMCID: PMC9288903 DOI: 10.7507/1002-1892.202203028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To construct three-dimensional (3D) pre-vascularized microstructures and explore the promoting effect of human fibroblasts (HFs) on the sprout and migration of human umbilical vein endothelial cells (HUVECs) in 3D co-culture system. METHODS HUVECs and HFs were cultured and the 3rd to 5th generation cells were selected for subsequent experiments. In 2D co-culture system, HFs were stained with PKH26 and the cell density was fixed, which co-cultured with HUVECs in different ratios (1∶4, 1∶1, 4∶1) and inoculation methods (HUVECs inoculation at 48 hours after HFs, direct mixed inoculation). Then the formation of vascular like structures was observed under fluorescence microscope. In 3D co-culture system, HUVECs and HFs were labeled with green fluorescent protein and red fluorescent protein by lentivirus transfection, respectively. They were inoculated on porous micro-carriers followed by dynamically culturing in rotating bottles to prepare HF, HUVEC, HF-EC, or HF&EC microstructures. The cell growth in microstructures was testing by low permeability crystal violet staining. Subsequently, the microstructures were embedded in fibrin gel and the cell growth and adhesion in HF and HUVEC microstructures were observed by laser confocal microscopy. Laser confocal microscope were also used to observe the sprouts of 4 kinds of microstructures, as well as the cell composition, the number and length of sprouts from HF-EC and HF&EC microstructures. HFs conditioned medium was prepared to observe its effect on sprouts of HUVEC microstructures with DMEM as control group. RESULTS In 2D co-culture system, HFs pre-culturing was helpful to the formation and stability of vascular like structures, and the best effect was when the ratio of two kinds of cells was 1∶1. In 3D co-culture system, it was found that the cells grew well on micro-carriers and had the ability of pre-vascularization. HUVEC microstructures did not sprout, but HF, HF-EC, and HF&EC microstructures could which indicated a good vascularization ability. The HF-EC microstructures were superior to HF&EC microstructures in terms of sprouts length and number ( P<0.05). The tubes sprouting from co-cultured group were composed of HFs and HUVECs, and HF microstructures migration preceded HUVEC microstructures always, and their migration trajectories were the same. HUVEC microstructures could sprout when cultured in HFs conditioned media. CONCLUSION HF-HUVEC pre-vascularized microstructures can be prepared by pre-culturing HFs before HUVECs and with the cell ratio at 1∶1 in a rotating bottle. In 3D co-culture system, HFs can promote and guide the sprout of HUVECs.
Collapse
|
13
|
Gene Expression Profile of Stromal Factors in Cancer-Associated Fibroblasts from Prostate Cancer. Diagnostics (Basel) 2022; 12:diagnostics12071605. [PMID: 35885510 PMCID: PMC9325062 DOI: 10.3390/diagnostics12071605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/07/2022] [Accepted: 06/22/2022] [Indexed: 11/26/2022] Open
Abstract
Recent investigations point at the stromal microenvironment to assess additional diagnostic information and provide new therapeutic targets in cancer. The aim of the study was to contribute to the characterization of the phenotype of cancer-associated fibroblasts (CAFs) in prostate cancer (PCa) compared with normal prostate-associated fibroblasts (NAFs) and fibroblasts from benign prostatic hyperplasia (BPH). Three patient populations were prospectively recruited: 23 patients with new localized PCa, 14 patients with advanced PCa treated with androgenic deprivation therapy (ADT), and 7 patients with BPH. Gene expression of 20 stroma-derived factors, including the androgen receptor (AR), chaperones (HSPA1A and HSF1), growth factors (FGF2, FGF7, FGF10, HGF, PDGFB, and TGFβ), proteins implicated in invasion (MMP2, MMP9, and MMP11), inflammation (IL6, IL17RB, NFκB, and STAT3), and in-stroma/epithelium interaction (CDH11, CXCL12, CXCL14, and FAP), was evaluated. Localized PCa CAFs showed a significant higher expression of FGF7, IL6, MMP2, and MMP11 compared with NAFs or IL17RB compared with BPH fibroblasts, but significantly lower expression of FGF10 and IL17RB compared with NAFs or CXCL14 compared with BPH fibroblasts. In addition, CAFs from ADT-resistant PCa showed significantly higher MMP11 and NFκB but significant lower TGFβ expression compared with CAFs from ADT-sensitive tumors. Our results contribute to defining the CAFs phenotypes associated to PCa progression, which may contribute to the diagnosis and design of alternative therapies in PCa.
Collapse
|
14
|
Wessolly M, Mairinger E, Borchert S, Bankfalvi A, Mach P, Schmid KW, Kimmig R, Buderath P, Mairinger FD. CAF-Associated Paracrine Signaling Worsens Outcome and Potentially Contributes to Chemoresistance in Epithelial Ovarian Cancer. Front Oncol 2022; 12:798680. [PMID: 35311102 PMCID: PMC8927667 DOI: 10.3389/fonc.2022.798680] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/07/2022] [Indexed: 01/06/2023] Open
Abstract
Background High-grade serous ovarian cancer (HGSOC) is the predominant and deadliest form of ovarian cancer. Some of its histological subtypes can be distinguished by frequent occurrence of cancer-associated myofibroblasts (CAFs) and desmoplastic stroma reaction (DSR). In this study, we want to explore the relationship between therapy outcome and the activity of CAF-associated signaling pathways in a homogeneous HGSOC patient collective. Furthermore, we want to validate these findings in a general Epithelial ovarian cancer (EOC) cohort. Methods The investigation cohort consists of 24 HGSOC patients. All of them were treated with platinum-based components and clinical follow-up was available. The validation cohort was comprised of 303 patients. Sequencing data (whole transcriptome) and clinical data were extracted from The Cancer Genome Atlas (TCGA). RNA of HGSOC patients was isolated using a Maxwell RSC instrument and the appropriate RNA isolation kit. For digital expression analysis a custom-designed gene panel was employed. All genes were linked to various DSR- and CAF- associated pathways. Expression analysis was performed on the NanoString nCounter platform. Finally, data were explored using the R programming environment (v. 4.0.3). Result In total, 15 CAF-associated genes were associated with patients’ survival. More specifically, 6 genes (MMP13, CGA, EPHA3, PSMD9, PITX2, PHLPP1) were linked to poor therapy outcome. Though a variety of different pathways appeared to be associated with therapy failure, many were related to CAF paracrine signaling, including MAPK, Ras and TGF-β pathways. Similar results were obtained from the validation cohort. Discussion In this study, we could successfully link CAF-associated pathways, as shown by increased Ras, MAPK and PI3K-Akt signaling to therapy failure (chemotherapy) in HGSOC and EOCs in general. As platinum-based chemotherapy has been the state-of-the-art therapy to treat HGSOC for decades, it is necessary to unveil the reasons behind resistance developments and poor outcome. In this work, CAF-associated signaling is shown to compromise therapy response. In the validation cohort, CAF-associated signaling is also associated with therapy failure in general EOC, possibly hinting towards a conserved mechanism. Therefore, it may be helpful to stratify HGSOC patients for CAF activity and consider alternative treatment options.
Collapse
Affiliation(s)
- Michael Wessolly
- Institute of Pathology, University Hospital Essen, Essen, Germany
- *Correspondence: Michael Wessolly,
| | - Elena Mairinger
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Sabrina Borchert
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Agnes Bankfalvi
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Pawel Mach
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | | | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Paul Buderath
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
15
|
Tissue engineering in dermatology - from lab to market. Tissue Cell 2022; 74:101717. [PMID: 34973574 DOI: 10.1016/j.tice.2021.101717] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/11/2021] [Accepted: 12/19/2021] [Indexed: 11/24/2022]
Abstract
Tissue Engineering is a branch of regenerative medical technology which helps replace damaged tissue using appropriate scaffolding, living cells, and growth factors. Using tissue engineering products can be a promising method for treating skin lesions such as wounds and deep burns. The interaction and interconnection of cells within the bio-culture medium or within a three-dimensional scaffold provides the conditions for tissue regeneration and subsequent healing of skin wounds. Tissue engineering in the field of dermatology has evolved over time from a single application of skin cells or biopolymer scaffolds to the use of cell and scaffold combinations for the treatment, repair, and closure of acute and chronic skin wounds. It has evolved. This technology has reached a point where most products are accepted, and the body rejects a small number, which strengthens the tissue engineering market. In this article, we aimed to review and study the market of this field by reviewing various articles on tissue engineering in the field of dermatology. Tissue-engineered skin substitutes are future options for wound healing and tissue regeneration strategies.
Collapse
|
16
|
Farrokhi A, Rahavi M, Jo S, Jalili R, Lim CJ, Ghahsary A, Reid GSD. Inflammatory Immune Responses Trigger Rejection of Allogeneic Fibroblasts Transplanted into Mouse Skin. Cell Transplant 2022; 31:9636897221113803. [PMID: 35912954 PMCID: PMC9340901 DOI: 10.1177/09636897221113803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fibroblasts, or their homolog stromal cells, are present in most tissues and play an essential role in tissue homeostasis and regeneration. As a result, fibroblast-based strategies have been widely employed in tissue engineering. However, while considered to have immunosuppressive properties, the survival and functionality of allogeneic fibroblasts after transplantation remain controversial. Here, we evaluated innate and adaptive immune responses against allogeneic fibroblasts following intradermal injection into different immune-deficient mouse strains. While allogeneic fibroblasts were rejected 1 week after transplantation in immunocompetent mice, rejection did not occur in immunodeficient γ chain–deficient NOD-SCID (NSG) mice. T-cell- and B-cell-deficient RAG1 knockout mice showed greater loss of fibroblasts by day 5 after transplantation compared with NSG mice (P ≤ 0.05) but prolonged persistence compared with wild-type recipient (P ≤ 0.005). Loss of fibroblasts correlated with the expression of proinflammatory chemokine genes and infiltration of myeloid cells in the transplantation site. Depletion of macrophages and neutrophils delayed rejection, revealing the role of innate immune cells in an early elimination of fibroblasts that is followed by T-cell-mediated rejection in the second week. These findings indicate that the application of allogeneic fibroblasts in tissue engineering products requires further improvements to overcome cell rejection by innate and adaptive immune cells.
Collapse
Affiliation(s)
- Ali Farrokhi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - MohammadReza Rahavi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Sumin Jo
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Reza Jalili
- Burn & Wound Healing Research Group, Division of Plastic Surgery, Department of Surgery and International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
| | - C. James Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - Aziz Ghahsary
- Burn & Wound Healing Research Group, Division of Plastic Surgery, Department of Surgery and International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
| | - Gregor S. D. Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Shafiee S, Shariatzadeh S, Zafari A, Majd A, Niknejad H. Recent Advances on Cell-Based Co-Culture Strategies for Prevascularization in Tissue Engineering. Front Bioeng Biotechnol 2021; 9:745314. [PMID: 34900955 PMCID: PMC8655789 DOI: 10.3389/fbioe.2021.745314] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the fabrication of a functional vascular network to maintain the viability of engineered tissues is a major bottleneck in the way of developing a more advanced engineered construct. Inspired by vasculogenesis during the embryonic period, the in vitro prevascularization strategies have focused on optimizing communications and interactions of cells, biomaterial and culture conditions to develop a capillary-like network to tackle the aforementioned issue. Many of these studies employ a combination of endothelial lineage cells and supporting cells such as mesenchymal stem cells, fibroblasts, and perivascular cells to create a lumenized endothelial network. These supporting cells are necessary for the stabilization of the newly developed endothelial network. Moreover, to optimize endothelial network development without impairing biomechanical properties of scaffolds or differentiation of target tissue cells, several other factors, including target tissue, endothelial cell origins, the choice of supporting cell, culture condition, incorporated pro-angiogenic factors, and choice of biomaterial must be taken into account. The prevascularization method can also influence the endothelial lineage cell/supporting cell co-culture system to vascularize the bioengineered constructs. This review aims to investigate the recent advances on standard cells used in in vitro prevascularization methods, their co-culture systems, and conditions in which they form an organized and functional vascular network.
Collapse
Affiliation(s)
- Sepehr Shafiee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Zafari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Majd
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Anitua E, Zalduendo M, Troya M, Tierno R, Alkhraisat MH. The inclusion of leukocytes into platelet rich plasma reduces scaffold stability and hinders extracellular matrix remodelling. Ann Anat 2021; 240:151853. [PMID: 34767933 DOI: 10.1016/j.aanat.2021.151853] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/14/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Scaffolds should have controllable degradation rate and allow cells to produce their own extracellular matrix. Platelet rich plasma (PRP) is a source of autologous growth factors and proteins embedded in a 3D fibrin scaffold. There is no consensus regarding the obtaining conditions and composition of PRPs. The aim of this study was to evaluate how the inclusion of leukocytes (L-PRP) in plasma rich in growth factors (PRGF) may alter the process of fibrinolysis. The effect of different combinations of cellular phenotypes with PRGF and L-PRP clots on both the fibrinolysis and matrix deposition process was also determined. METHODS PRGF and L-PRP clots were incubated for 14 days and D-dimer and type I collagen were determined in their conditioned media to evaluate clots' stability. For remodelling assays, gingival fibroblasts, alveolar osteoblasts and human umbilical vein endothelial cells (HUVEC) were seeded onto the two types of clots for 14 days. D-dimer, type I collagen, and laminin α4 were measured by ELISA kits in their conditioned media. Morphological and histological analysis were also performed. Cell proliferation was additionally determined RESULTS: PRGF clots preserved their stability as shown by the low levels of both D-dimer and collagen type I compared to those obtained for L-PRP clots. The inclusion of both gingival fibroblasts and alveolar osteoblasts stimulated a higher fibrinolysis in the PRGF clots. In contrast to this, the degradation rates of both PRGF and L-PRP clots remained unchanged after culturing with the endothelial cells. In all cases, type I collagen and laminin α4 levels were in line with the degree of clots' degradation. In all phenotypes, cell proliferation was significantly higher in PRGF than in L-PRP clots. CONCLUSION The inclusion of leukocytes in PRGF scaffolds reduced their stability, decreased cell number and slowed down cell remodelling.
Collapse
Affiliation(s)
- Eduardo Anitua
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| | - Mar Zalduendo
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | | | - Roberto Tierno
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Mohammad H Alkhraisat
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| |
Collapse
|
19
|
Gurov AV, Kryukov AI, Levina JV, Bakhtin AA, Dubovaya TK, Murzakhanova ZV. [Hearing dynamics in acute traumatic perforation of the tympanic membrane after application of blood plasma enriched with platelet growth factors]. Vestn Otorinolaringol 2021; 86:23-30. [PMID: 34499443 DOI: 10.17116/otorino20218604123] [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] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The article considers the possibility of treating patients with acute traumatic perforation of the eardrum with a single application of blood plasma enriched with platelet-derived growth factors in the perforation area. As a result of the studies, it was found that the acceleration of its closure and the restoration of hearing. MATERIAL AND METHODS The study included 43 patients with unilateral acute PBP of various etiologies (Table 1). Patients were divided into 2 groups - the main - 24 patients (10 men and 14 women), the average age - 30±9.7 years and the control - 19 patients (9 men and 10 women), the average age - 26.5±6.37 years. The compared groups did not reliably differ in age, gender, frequency and severity of hearing impairment. RESULTS Analyzing the results, we can confidently talk about the beneficial effect of blood plasma enriched in platelet-derived growth factors on the speed, completeness of closure of traumatic perforation of the eardrum and the dynamics of restoration of auditory function. The use of blood plasma enriched with platelet growth factors led to a regular reduction in the time and increase in the frequency of cases of early closure of perforated eardrum, as well as improvement and restoration of auditory function according to the results of tonal threshold audiometry and otoacoustic emission in dynamics. CONCLUSION Being generally available even in outpatient practice, the method of a single application of blood plasma enriched with platelet growth factors in the area of perforation of the tympanic membrane can significantly accelerate the healing process of the tympanic membrane, while improving the quality of life of the patient.
Collapse
Affiliation(s)
- A V Gurov
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of Russia, Moscow, Russia.,L.I. Sverzhevsky Otorhinolaryngology Scientific Research Clinical Institute Moscow Health Department, Moscow, Russia
| | - A I Kryukov
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of Russia, Moscow, Russia.,L.I. Sverzhevsky Otorhinolaryngology Scientific Research Clinical Institute Moscow Health Department, Moscow, Russia
| | - Ju V Levina
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of Russia, Moscow, Russia.,L.I. Sverzhevsky Otorhinolaryngology Scientific Research Clinical Institute Moscow Health Department, Moscow, Russia
| | - A A Bakhtin
- The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia, Moscow, Russia
| | - T K Dubovaya
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of Russia, Moscow, Russia
| | - Z V Murzakhanova
- N.I. Pirogov Russian National Research Medical University of the Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
20
|
Hou J, Yang R, Vuong I, Li F, Kong J, Mao HQ. Biomaterials strategies to balance inflammation and tenogenesis for tendon repair. Acta Biomater 2021; 130:1-16. [PMID: 34082095 DOI: 10.1016/j.actbio.2021.05.043] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/15/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
Adult tendon tissue demonstrates a limited regenerative capacity, and the natural repair process leaves fibrotic scar tissue with inferior mechanical properties. Surgical treatment is insufficient to provide the mechanical, structural, and biochemical environment necessary to restore functional tissue. While numerous strategies including biodegradable scaffolds, bioactive factor delivery, and cell-based therapies have been investigated, most studies have focused exclusively on either suppressing inflammation or promoting tenogenesis, which includes tenocyte proliferation, ECM production, and tissue formation. New biomaterials-based approaches represent an opportunity to more effectively balance the two processes and improve regenerative outcomes from tendon injuries. Biomaterials applications that have been explored for tendon regeneration include formation of biodegradable scaffolds presenting topographical, mechanical, and/or immunomodulatory cues conducive to tendon repair; delivery of immunomodulatory or tenogenic biomolecules; and delivery of therapeutic cells such as tenocytes and stem cells. In this review, we provide the biological context for the challenges in tendon repair, discuss biomaterials approaches to modulate the immune and regenerative environment during the healing process, and consider the future development of comprehensive biomaterials-based strategies that can better restore the function of injured tendon. STATEMENT OF SIGNIFICANCE: Current strategies for tendon repair focus on suppressing inflammation or enhancing tenogenesis. Evidence indicates that regulated inflammation is beneficial to tendon healing and that excessive tissue remodeling can cause fibrosis. Thus, it is necessary to adopt an approach that balances the benefits of regulated inflammation and tenogenesis. By reviewing potential treatments involving biodegradable scaffolds, biological cues, and therapeutic cells, we contrast how each strategy promotes or suppresses specific repair steps to improve the healing outcome, and highlight the advantages of a comprehensive approach that facilitates the clearance of necrotic tissue and recruitment of cells during the inflammatory stage, followed by ECM synthesis and organization in the proliferative and remodeling stages with the goal of restoring function to the tendon.
Collapse
|
21
|
Masson-Meyers DS, Tayebi L. Vascularization strategies in tissue engineering approaches for soft tissue repair. J Tissue Eng Regen Med 2021; 15:747-762. [PMID: 34058083 DOI: 10.1002/term.3225] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Insufficient vascularization during tissue repair is often associated with poor clinical outcomes. This is a concern especially when patients have critical-sized injuries, where the size of the defect restricts vascularity, or even in small defects that have to be treated under special conditions, such as after radiation therapy (relevant to tumor resection) that hinders vascularity. In fact, poor vascularization is one of the major obstacles for clinical application of tissue engineering methods in soft tissue repair. As a key issue, lack of graft integration, caused by inadequate vascularization after implantation, can lead to graft failure. Moreover, poor vascularization compromises the viability of cells seeded in deep portions of scaffolds/graft materials, due to hypoxia and insufficient nutrient supply. In this article we aim to review vascularization strategies employed in tissue engineering techniques to repair soft tissues. For this purpose, we start by providing a brief overview of the main events during the physiological wound healing process in soft tissues. Then, we discuss how tissue repair can be achieved through tissue engineering, and considerations with regards to the choice of scaffold materials, culture conditions, and vascularization techniques. Next, we highlight the importance of vascularization, along with strategies and methods of prevascularization of soft tissue equivalents, particularly cell-based prevascularization. Lastly, we present a summary of commonly used in vitro methods during the vascularization of tissue-engineered soft tissue constructs.
Collapse
Affiliation(s)
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| |
Collapse
|
22
|
Ratri MC, Brilian AI, Setiawati A, Nguyen HT, Soum V, Shin K. Recent Advances in Regenerative Tissue Fabrication: Tools, Materials, and Microenvironment in Hierarchical Aspects. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000088] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Monica Cahyaning Ratri
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
- Department of Chemistry Education Sanata Dharma University Yogyakarta 55281 Indonesia
| | - Albertus Ivan Brilian
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Agustina Setiawati
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
- Department of Life Science Sogang University Seoul 04107 Republic of Korea
- Faculty of Pharmacy Sanata Dharma University Yogyakarta 55281 Indonesia
| | - Huong Thanh Nguyen
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Veasna Soum
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| |
Collapse
|
23
|
Richardson IM, Calo CJ, Hind LE. Microphysiological Systems for Studying Cellular Crosstalk During the Neutrophil Response to Infection. Front Immunol 2021; 12:661537. [PMID: 33986752 PMCID: PMC8111168 DOI: 10.3389/fimmu.2021.661537] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are the primary responders to infection, rapidly migrating to sites of inflammation and clearing pathogens through a variety of antimicrobial functions. This response is controlled by a complex network of signals produced by vascular cells, tissue resident cells, other immune cells, and the pathogen itself. Despite significant efforts to understand how these signals are integrated into the neutrophil response, we still do not have a complete picture of the mechanisms regulating this process. This is in part due to the inherent disadvantages of the most-used experimental systems: in vitro systems lack the complexity of the tissue microenvironment and animal models do not accurately capture the human immune response. Advanced microfluidic devices incorporating relevant tissue architectures, cell-cell interactions, and live pathogen sources have been developed to overcome these challenges. In this review, we will discuss the in vitro models currently being used to study the neutrophil response to infection, specifically in the context of cell-cell interactions, and provide an overview of their findings. We will also provide recommendations for the future direction of the field and what important aspects of the infectious microenvironment are missing from the current models.
Collapse
Affiliation(s)
| | | | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO, United States
| |
Collapse
|
24
|
Ruiz-Alonso S, Lafuente-Merchan M, Ciriza J, Saenz-Del-Burgo L, Pedraz JL. Tendon tissue engineering: Cells, growth factors, scaffolds and production techniques. J Control Release 2021; 333:448-486. [PMID: 33811983 DOI: 10.1016/j.jconrel.2021.03.040] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Tendon injuries are a global health problem that affects millions of people annually. The properties of tendons make their natural rehabilitation a very complex and long-lasting process. Thanks to the development of the fields of biomaterials, bioengineering and cell biology, a new discipline has emerged, tissue engineering. Within this discipline, diverse approaches have been proposed. The obtained results turn out to be promising, as increasingly more complex and natural tendon-like structures are obtained. In this review, the nature of the tendon and the conventional treatments that have been applied so far are underlined. Then, a comparison between the different tendon tissue engineering approaches that have been proposed to date is made, focusing on each of the elements necessary to obtain the structures that allow adequate regeneration of the tendon: growth factors, cells, scaffolds and techniques for scaffold development. The analysis of all these aspects allows understanding, in a global way, the effect that each element used in the regeneration of the tendon has and, thus, clarify the possible future approaches by making new combinations of materials, designs, cells and bioactive molecules to achieve a personalized regeneration of a functional tendon.
Collapse
Affiliation(s)
- Sandra Ruiz-Alonso
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain
| | - Markel Lafuente-Merchan
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain
| | - Jesús Ciriza
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Laura Saenz-Del-Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
| |
Collapse
|
25
|
Marinkovic M, Sridharan R, Santarella F, Smith A, Garlick JA, Kearney CJ. Optimization of extracellular matrix production from human induced pluripotent stem cell-derived fibroblasts for scaffold fabrication for application in wound healing. J Biomed Mater Res A 2021; 109:1803-1811. [PMID: 33755305 DOI: 10.1002/jbm.a.37173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/16/2021] [Accepted: 03/12/2021] [Indexed: 01/01/2023]
Abstract
Extracellular matrix is a key component of all tissues, including skin and it plays a crucial role in the complex events of wound healing. These events are impaired in chronic wounds, with chronic inflammation and infection often present in these non-healing wounds. Many tissue engineering approaches for wound healing provide a scaffold to mimic the native matrix. Fibroblasts derived from iPS cells (iPSF) represent a novel source of matrix rich in pro-regenerative components, which can be used for scaffold fabrication to improve wound healing. However, in vitro production of matrix by cells for scaffold fabrication requires long cell culturing times which increases cost. The aim of this work is to optimize the iPSF matrix production by boosting matrix deposition, without affecting its composition. A good candidate technique to achieve this goal is macromolecular crowding, which is known to promote conversion of procollagen into mature collagen and its accumulation. We tested two molecular crowders, Ficoll and Carrageenan-in combination with ascorbic acid-over a prolonged period of time. Ficoll in combination with ascorbic acid notably increased collagen deposition and matrix dry weight compared to ascorbic acid alone, and did not affect matrix composition as measured by RT-PCR. Interestingly, Carrageenan did not affect collagen quantity, but it significantly increased glycosaminoglycan deposition. Finally, we successfully fabricated scaffolds from harvested matrix and confirmed their ability for cell growth and viability. This work lays the foundation for development of a time and cost effective protocol for novel iPSF ECM production for tissue engineering scaffolds.
Collapse
Affiliation(s)
- Milica Marinkovic
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland
| | - Rukmani Sridharan
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland
| | - Francesco Santarella
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland
| | - Avi Smith
- Garlick Lab, Department of Diagnostic Sciences, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Jonathan A Garlick
- Garlick Lab, Department of Diagnostic Sciences, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Cathal J Kearney
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland.,AMBER Research Center, Trinity College Dublin & RCSI and Trinity Centre for Bioengineering, Dublin 2, Ireland.,Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
26
|
Pita-López ML, Fletes-Vargas G, Espinosa-Andrews H, Rodríguez-Rodríguez R. Physically cross-linked chitosan-based hydrogels for tissue engineering applications: A state-of-the-art review. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110176] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Fernández Muñoz B, Lopez-Navas L, Gonzalez Bermejo M, Lomas Romero IM, Montiel Aguilera MÁ, Campos Cuerva R, Arribas Arribas B, Nogueras S, Carmona Sánchez G, Santos González M. A PROPRIETARY GMP HUMAN PLATELET LYSATE FOR THE EXPANSION OF DERMAL FIBROBLASTS FOR CLINICAL APPLICATIONS. Platelets 2021; 33:98-109. [PMID: 33393414 DOI: 10.1080/09537104.2020.1856356] [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] [Indexed: 01/01/2023]
Abstract
Recent years have witnessed the introduction of ex vivo expanded dermal fibroblasts for several cell therapy and tissue-engineering applications, including the treatment of facial scars and burns, representing a promising cell type for regenerative medicine. We tested different in-house produced human platelet lysate (HPL) solutions against fetal bovine serum as supplements for in vitro fibroblast expansion by comparing cell yield, molecular marker expression, extracellular matrix (ECM) generation, genomic stability and global gene expression. Our in-house produced HPL supported fibroblast growth at levels similar to those for FBS and commercial HPL products and was superior to AB human serum. Cells grown in HPL maintained a fibroblast phenotype (VIM+, CD44+, CD13+, CD90+), ECM generation capacity (FN+, COL1+) and a normal karyotype, although gene expression profiling revealed changes related to cell metabolism, adhesion and cellular senescence. The HPL manufacturing process was validated within a GMP compliant system and the solution was stable at -80ºC and -20ºC for 2 years. Dermal fibroblasts expanded in vitro with HPL maintain a normal karyotype and expression of fibroblast markers, with only minor changes in their global gene expression profile. Our in-house produced GMP-HPL is an efficient, safe and economical cell culture supplement that can help increase the healthcare activity of blood transfusion centers through the re-use of transfusional plasma and platelets approaching their expiration date. Currently, our HPL solution is approved by the Spanish Agency of Medicines and Medical Devices and is being used in the manufacture of cell therapy products.
Collapse
Affiliation(s)
- Beatriz Fernández Muñoz
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Departamento de Neurociencia Aplicada, Instituto de Investigaciones Biomédicas de Sevilla (IBIS), Seville, Spain
| | - Luis Lopez-Navas
- Unidad de Coordinación, Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain
| | - María Gonzalez Bermejo
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Biología Molecular, Biomedicina e Investigación Clínica, University of Seville, Seville, Spain
| | - Isabel María Lomas Romero
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain
| | - Miguel Ángel Montiel Aguilera
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain
| | - Rafael Campos Cuerva
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Biología Molecular, Biomedicina e Investigación Clínica, University of Seville, Seville, Spain.,Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Fundación Pública Andaluza para la Gestión de la Investigación en Salud en Sevilla (FISEVI), Seville, Spain
| | - Blanca Arribas Arribas
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Pharmaceutical Technology and Medicine Sciences (Pharmacy), University of Seville, Seville, Spain
| | - Sonia Nogueras
- Departamento de Terapia Celular, Instituto Maimónides de Investigación Biomédica of Córdoba (IMIBIC), Córdoba, Spain.,Unidad de Terapia Celular, Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Gloria Carmona Sánchez
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Unidad de Coordinación, Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Biomedicine, University of Granada, Granada, Spain
| | - Mónica Santos González
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Fundación Pública Andaluza para la Gestión de la Investigación en Salud en Sevilla (FISEVI), Seville, Spain
| |
Collapse
|
28
|
Chen AX, Chhabra A, Song HHG, Fleming HE, Chen CS, Bhatia SN. Controlled Apoptosis of Stromal Cells to Engineer Human Microlivers. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910442. [PMID: 33776613 PMCID: PMC7996305 DOI: 10.1002/adfm.201910442] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Engineered tissue models comprise a variety of multiplexed ensembles in which combinations of epithelial, stromal, and immune cells give rise to physiologic function. Engineering spatiotemporal control of cell-cell and cell-matrix interactions within these 3D multicellular tissues would represent a significant advance for tissue engineering. In this work, a new method, entitled CAMEO (Controlled Apoptosis in Multicellular tissues for Engineered Organogenesis) enables the non-invasive triggering of controlled apoptosis to eliminate genetically-engineered cells from a pre-established culture. Using this approach, the contribution of stromal cells to the phenotypic stability of primary human hepatocytes is examined. 3D hepatic microtissues, in which fibroblasts can enhance phenotypic stability and accelerate aggregation into spheroids, were found to rely only transiently on fibroblast interaction to support multiple axes of liver function, such as protein secretion and drug detoxification. Due to its modularity, CAMEO has the promise to be readily extendable to other applications that are tied to the complexity of 3D tissue biology, from understanding in vitro organoid models to building artificial tissue grafts.
Collapse
Affiliation(s)
- Amanda X Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnav Chhabra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - H-H Greco Song
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather E Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
29
|
Song HHG, Lammers A, Sundaram S, Rubio L, Chen AX, Li L, Eyckmans J, Bhatia SN, Chen CS. Transient Support from Fibroblasts is Sufficient to Drive Functional Vascularization in Engineered Tissues. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2003777. [PMID: 33613149 PMCID: PMC7891457 DOI: 10.1002/adfm.202003777] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 05/05/2023]
Abstract
Formation of capillary blood vasculature is a critical requirement for native as well as engineered organs and can be induced in vitro by co-culturing endothelial cells with fibroblasts. However, whether these fibroblasts are required only in the initial morphogenesis of endothelial cells or needed throughout is unknown, and the ability to remove these stromal cells after assembly could be useful for clinical translation. In this study, we introduce a technique termed CAMEO (Controlled Apoptosis in Multicellular Tissues for Engineered Organogenesis), whereby fibroblasts are selectively ablated on demand, and utilize it to probe the dispensability of fibroblasts in vascular morphogenesis. The presence of fibroblasts is shown to be necessary only during the first few days of endothelial cell morphogenesis, after which they can be ablated without significantly affecting the structural and functional features of the developed vasculature. Furthermore, we demonstrate the use of CAMEO to vascularize a construct containing primary human hepatocytes that improved tissue function. In conclusion, this study suggests that transient, initial support from fibroblasts is sufficient to drive vascular morphogenesis in engineered tissues, and this strategy of engineering-via-elimination may provide a new general approach for achieving desired functions and cell compositions in engineered organs.
Collapse
Affiliation(s)
- H-H Greco Song
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alex Lammers
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Subramanian Sundaram
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Logan Rubio
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Amanda X Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Linqing Li
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jeroen Eyckmans
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sangeeta N Bhatia
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
30
|
Sayers JR, Riley PR. Heart regeneration: beyond new muscle and vessels. Cardiovasc Res 2020; 117:727-742. [PMID: 33241843 DOI: 10.1093/cvr/cvaa320] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/16/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
The most striking consequence of a heart attack is the loss of billions of heart muscle cells, alongside damage to the associated vasculature. The lost cardiovascular tissue is replaced by scar formation, which is non-functional and results in pathological remodelling of the heart and ultimately heart failure. It is, therefore, unsurprising that the heart regeneration field has centred efforts to generate new muscle and blood vessels through targeting cardiomyocyte proliferation and angiogenesis following injury. However, combined insights from embryological studies and regenerative models, alongside the adoption of -omics technology, highlight the extensive heterogeneity of cell types within the forming or re-forming heart and the significant crosstalk arising from non-muscle and non-vessel cells. In this review, we focus on the roles of fibroblasts, immune, conduction system, and nervous system cell populations during heart development and we consider the latest evidence supporting a function for these diverse lineages in contributing to regeneration following heart injury. We suggest that the emerging picture of neurologically, immunologically, and electrically coupled cell function calls for a wider-ranging combinatorial approach to heart regeneration.
Collapse
Affiliation(s)
- Judy R Sayers
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
31
|
LeBleu VS, Neilson EG. Origin and functional heterogeneity of fibroblasts. FASEB J 2020; 34:3519-3536. [PMID: 32037627 DOI: 10.1096/fj.201903188r] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
The inherent plasticity and resiliency of fibroblasts make this cell type a conventional tool for basic research. But where do they come from, are all fibroblasts the same, and how do they function in disease? The first fibroblast lineages in mammalian development emerge from the ooze of primary mesenchyme during gastrulation. They are cells that efficiently create and negotiate the extracellular matrix of the mesoderm in order to migrate and meet their developmental fate. Mature fibroblasts in epithelial tissues live in the interstitial spaces between basement membranes that spatially delimit complex organ structures. While the function of resident fibroblasts in healthy tissues is largely conjecture, the accumulation of fibroblasts in pathologic lesions offers insight into biologic mechanisms that control their function; fibroblasts are poised to coordinate fibrogenesis in tissue injury, neoplasia, and aging. Here, we examine the developmental origin and plasticity of fibroblasts, their molecular and functional definitions, the epigenetic control underlying their identity and activation, and the evolution of their immune regulatory functions. These topics are reviewed through the lens of fate mapping using genetically engineered mouse models and from the perspective of single-cell RNA sequencing. Recent observations suggest dynamic and heterogeneous functions for fibroblasts that underscore their complex molecular signatures and utility in injured tissues.
Collapse
Affiliation(s)
- Valerie S LeBleu
- Departments of Medicine and Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eric G Neilson
- Departments of Medicine and Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
32
|
Camasão DB, González-Pérez M, Palladino S, Alonso M, Rodríguez-Cabello JC, Mantovani D. Elastin-like recombinamers in collagen-based tubular gels improve cell-mediated remodeling and viscoelastic properties. Biomater Sci 2020; 8:3536-3548. [DOI: 10.1039/d0bm00292e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The partial substitution of collagen with elastin-like recombinamers in tubular gels improves cell-mediated remodeling, elastic moduli and strength during maturation.
Collapse
Affiliation(s)
- Dimitria Bonizol Camasão
- Laboratory for Biomaterials and Bioengineering
- Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery
- Department of Min-Met-Materials Engineering
- Research Center of CHU de Québec
- Division of Regenerative Medicine
| | - Miguel González-Pérez
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology)
- CIBER-BBN
- University of Valladolid
- 47011 Valladolid
- Spain
| | - Sara Palladino
- Laboratory for Biomaterials and Bioengineering
- Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery
- Department of Min-Met-Materials Engineering
- Research Center of CHU de Québec
- Division of Regenerative Medicine
| | - Matilde Alonso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology)
- CIBER-BBN
- University of Valladolid
- 47011 Valladolid
- Spain
| | | | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering
- Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery
- Department of Min-Met-Materials Engineering
- Research Center of CHU de Québec
- Division of Regenerative Medicine
| |
Collapse
|
33
|
Urciuolo F, Casale C, Imparato G, Netti PA. Bioengineered Skin Substitutes: the Role of Extracellular Matrix and Vascularization in the Healing of Deep Wounds. J Clin Med 2019; 8:E2083. [PMID: 31805652 PMCID: PMC6947552 DOI: 10.3390/jcm8122083] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
The formation of severe scars still represents the result of the closure process of extended and deep skin wounds. To address this issue, different bioengineered skin substitutes have been developed but a general consensus regarding their effectiveness has not been achieved yet. It will be shown that bioengineered skin substitutes, although representing a valid alternative to autografting, induce skin cells in repairing the wound rather than guiding a regeneration process. Repaired skin differs from regenerated skin, showing high contracture, loss of sensitivity, impaired pigmentation and absence of cutaneous adnexa (i.e., hair follicles and sweat glands). This leads to significant mobility and aesthetic concerns, making the development of more effective bioengineered skin models a current need. The objective of this review is to determine the limitations of either commercially available or investigational bioengineered skin substitutes and how advanced skin tissue engineering strategies can be improved in order to completely restore skin functions after severe wounds.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; (C.C.); (P.A.N.)
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II P.le Tecchio 80, 80125 Naples, Italy
| | - Costantino Casale
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; (C.C.); (P.A.N.)
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy;
| | - Paolo A. Netti
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; (C.C.); (P.A.N.)
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II P.le Tecchio 80, 80125 Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy;
| |
Collapse
|
34
|
What Is the Fuss about Integrins and the Tumor Microenvironment? Cancers (Basel) 2019; 11:cancers11091296. [PMID: 31484335 PMCID: PMC6770914 DOI: 10.3390/cancers11091296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 01/17/2023] Open
|
35
|
Zeltz C, Primac I, Erusappan P, Alam J, Noel A, Gullberg D. Cancer-associated fibroblasts in desmoplastic tumors: emerging role of integrins. Semin Cancer Biol 2019; 62:166-181. [PMID: 31415910 DOI: 10.1016/j.semcancer.2019.08.004] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) is a complex meshwork of extracellular matrix (ECM) macromolecules filled with a collection of cells including cancer-associated fibroblasts (CAFs), blood vessel associated smooth muscle cells, pericytes, endothelial cells, mesenchymal stem cells and a variety of immune cells. In tumors the homeostasis governing ECM synthesis and turnover is disturbed resulting in abnormal blood vessel formation and excessive fibrillar collagen accumulations of varying stiffness and organization. The disturbed ECM homeostasis opens up for new types of paracrine, cell-cell and cell-ECM interactions with large consequences for tumor growth, angiogenesis, metastasis, immune suppression and resistance to treatments. As a main producer of ECM and paracrine signals the CAF is a central cell type in these events. Whereas the paracrine signaling has been extensively studied in the context of tumor-stroma interactions, the nature of the numerous integrin-mediated cell-ECM interactions occurring in the TME remains understudied. In this review we will discuss and dissect the role of known and potential CAF interactions in the TME, during both tumorigenesis and chemoresistance-induced events, with a special focus on the "interaction landscape" in desmoplastic breast, lung and pancreatic cancers. As an example of the multifaceted mode of action of the stromal collagen receptor integrin α11β1, we will summarize our current understanding on the role of this CAF-expressed integrin in these three tumor types.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway; Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Irina Primac
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liege (ULiège), Liege, Belgium
| | - Pugazendhi Erusappan
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway; Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Jahedul Alam
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Agnes Noel
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liege (ULiège), Liege, Belgium
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway.
| |
Collapse
|
36
|
Calejo I, Costa-Almeida R, Reis RL, Gomes ME. Enthesis Tissue Engineering: Biological Requirements Meet at the Interface. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:330-356. [DOI: 10.1089/ten.teb.2018.0383] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Isabel Calejo
- 3B's Research Group, I3Bs—Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Raquel Costa-Almeida
- 3B's Research Group, I3Bs—Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs—Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's—PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Center for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Manuela E. Gomes
- 3B's Research Group, I3Bs—Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's—PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Center for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| |
Collapse
|
37
|
Costa-Almeida R, Calejo I, Gomes ME. Mesenchymal Stem Cells Empowering Tendon Regenerative Therapies. Int J Mol Sci 2019; 20:E3002. [PMID: 31248196 PMCID: PMC6627139 DOI: 10.3390/ijms20123002] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 12/19/2022] Open
Abstract
Tendon tissues have limited healing capacity. The incidence of tendon injuries and the unsatisfactory functional outcomes of tendon repair are driving the search for alternative therapeutic approaches envisioning tendon regeneration. Cellular therapies aim at delivering adequate, regeneration-competent cell types to the injured tendon and toward ultimately promoting its reconstruction and recovery of functionality. Mesenchymal stem cells (MSCs) either obtained from tendons or from non-tendon sources, like bone marrow (BM-MSCs) or adipose tissue (ASCs), have been receiving increasing attention over the years toward enhancing tendon healing. Evidences from in vitro and in vivo studies suggest MSCs can contribute to accelerate and improve the quality of tendon healing. Nonetheless, the exact mechanisms underlying these repair events are yet to be fully elucidated. This review provides an overview of the main challenges in the field of cell-based regenerative therapies, discussing the role of MSCs in boosting tendon regeneration, particularly through their capacity to enhance the tenogenic properties of tendon resident cells.
Collapse
Affiliation(s)
- Raquel Costa-Almeida
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Barco, Guimarães, Portugal.
| | - Isabel Calejo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Barco, Guimarães, Portugal.
| | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4805-017 Barco, Guimarães, Portugal.
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal.
| |
Collapse
|
38
|
Berndt S, Turzi A, Pittet-Cuénod B, Modarressi A. Autologous Platelet-Rich Plasma (CuteCell PRP) Safely Boosts In Vitro Human Fibroblast Expansion. Tissue Eng Part A 2019; 25:1550-1563. [PMID: 30896295 DOI: 10.1089/ten.tea.2018.0335] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Nowadays autologous fibroblast application for skin repair presents an important clinical interest. In most cases, in vitro skin cell culture is mandatory. However, cell expansion using xenogeneic or allogenic culture media presents some disadvantages, such as the risk of infection transmission or slow cell expansion. In this study, we investigated an autologous culture system to expand human skin fibroblast cells in vitro with the patient's own platelet-rich plasma (PRP). Human dermal fibroblasts were isolated from patients undergoing abdominoplasty, and blood was collected to prepare nonactivated PRP using the CuteCell™ PRP medical device. Cultures were followed up to 7 days using a medium supplemented with either fetal bovine serum (FBS) or PRP. Fibroblasts cultured in medium supplemented with PRP showed dose-dependently significantly higher proliferation rates (up to 7.7 times with 20% of PRP) and initiated a faster migration in the in vitro wound healing assay compared with FBS, while chromosomal stability was maintained. At high concentrations, PRP changed fibroblast morphology, inducing cytoskeleton rearrangement and an increase of alpha-smooth muscle actin and vimentin expression. Our findings show that autologous PRP is an efficient and cost-effective supplement for fibroblast culture, and should be considered as a safe alternative to xenogeneic/allogenic blood derivatives for in vitro cell expansion. Impact Statement Autologous dermal fibroblast graft is an important therapy in skin defect repair, but in vitro skin cell culture is mandatory in most cases. However, cell expansion using xenogeneic/allogenic culture media presents some disadvantages, such as the risk of infection transmission. We demonstrated that an autologous culture system with the patient's own platelet-rich plasma is an efficient, cost-effective, and safe supplement for fibroblast culture. As it respects the good manufacturing practices and regulatory agencies standards, it should be considered as a potent alternative and substitute to xenogeneic or allogenic blood derivatives for the validation of future clinical protocols using in vitro cell expansion.
Collapse
Affiliation(s)
- Sarah Berndt
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland.,Regen Lab SA, Le Mont-sur-Lausanne, Switzerland
| | | | - Brigitte Pittet-Cuénod
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Ali Modarressi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
39
|
Campos-Cuerva R, Fernández-Muñoz B, Farfán López F, Pereira Arenas S, Santos-González M, Lopez-Navas L, Alaminos M, Campos A, Muntané J, Cepeda-Franco C, Gómez-Bravo MÁ. Nanostructured fibrin agarose hydrogel as a novel haemostatic agent. J Tissue Eng Regen Med 2019; 13:664-673. [PMID: 30793853 PMCID: PMC6594136 DOI: 10.1002/term.2831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/12/2019] [Accepted: 02/21/2019] [Indexed: 12/18/2022]
Abstract
Blood loss remains a major concern during surgery and can increase the morbidity of the intervention. The use of topical haemostatic agents to overcome this issue therefore becomes necessary. Fibrin sealants are promising haemostatic agents due to their capacity to promote coagulation, but their effectiveness and applicability need to be improved. We have compared the haemostatic efficacy of a novel nanostructured fibrin‐agarose hydrogel patch, with (c‐NFAH) or without cells (a‐NFAH), against two commercially available haemostatic agents in a rat model of hepatic resection. Hepatic resections were performed by making short or long incisions (mild or severe model, respectively), and haemostatic agents were applied to evaluate time to haemostasis, presence of haematoma, post‐operative adhesions to adjacent tissues, and inflammation factors. We found a significantly higher haemostatic success rate (time to haemostasis) with a‐NFAH than with other commercial haemostatic agents. Furthermore, other relevant outcomes investigated were also improved in the a‐NFAH group, including no presence of haematoma, lower adhesions, and lower grades of haemorrhage, inflammation, and necrosis in histological analysis. Overall, these findings identify a‐NFAH as a promising haemostatic agent in liver resection and likely in a range of surgical procedures.
Collapse
Affiliation(s)
- Rafael Campos-Cuerva
- Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Seville, Spain.,Cell Therapy and Cell Reprogramming Unit, GMP Network of the Andalusian Initiative for Advanced Therapies, Junta de Andalucia, Seville, Spain.,PhD Program in Molecular Biology, Biomedicine and Clinical Research, Universidad de Sevilla, 41013, Seville, Spain
| | - Beatriz Fernández-Muñoz
- Cell Therapy and Cell Reprogramming Unit, GMP Network of the Andalusian Initiative for Advanced Therapies, Junta de Andalucia, Seville, Spain.,Instituto de Biomedicina de Sevilla, IBIS (HUVR/CSIC/Universidad de Sevilla, Junta de Andalucía), Spain
| | - Francisco Farfán López
- Servicio de Anatomía Patológica, Hospital Universitario Virgen del Rocío (HUVR), Seville, Spain
| | - Sheila Pereira Arenas
- Instituto de Biomedicina de Sevilla, IBIS (HUVR/CSIC/Universidad de Sevilla, Junta de Andalucía), Spain
| | - Mónica Santos-González
- Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Seville, Spain.,Cell Therapy and Cell Reprogramming Unit, GMP Network of the Andalusian Initiative for Advanced Therapies, Junta de Andalucia, Seville, Spain
| | - Luis Lopez-Navas
- Andalusian Initiative for Advanced Therapies, Junta de Andalucia, Seville, Spain
| | - Miguel Alaminos
- Department of Histology, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, ibs.GRANADA, 18016 Granada, Spain
| | - Antonio Campos
- Department of Histology, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, ibs.GRANADA, 18016 Granada, Spain
| | - Jordi Muntané
- Instituto de Biomedicina de Sevilla, IBIS (HUVR/CSIC/Universidad de Sevilla, Junta de Andalucía), Spain.,Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Carmen Cepeda-Franco
- Instituto de Biomedicina de Sevilla, IBIS (HUVR/CSIC/Universidad de Sevilla, Junta de Andalucía), Spain.,Unidad de Cirugia Hepato-Bilio-Pancreática y Trasplantes, HUVR, Spain
| | - Miguel Ángel Gómez-Bravo
- Instituto de Biomedicina de Sevilla, IBIS (HUVR/CSIC/Universidad de Sevilla, Junta de Andalucía), Spain.,Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain.,Unidad de Cirugia Hepato-Bilio-Pancreática y Trasplantes, HUVR, Spain
| |
Collapse
|
40
|
Witherel CE, Abebayehu D, Barker TH, Spiller KL. Macrophage and Fibroblast Interactions in Biomaterial-Mediated Fibrosis. Adv Healthc Mater 2019; 8:e1801451. [PMID: 30658015 PMCID: PMC6415913 DOI: 10.1002/adhm.201801451] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/07/2018] [Indexed: 01/08/2023]
Abstract
Biomaterial-mediated inflammation and fibrosis remain a prominent challenge in designing materials to support tissue repair and regeneration. Despite the many biomaterial technologies that have been designed to evade or suppress inflammation (i.e., delivery of anti-inflammatory drugs, hydrophobic coatings, etc.), many materials are still subject to a foreign body response, resulting in encapsulation of dense, scar-like extracellular matrix. The primary cells involved in biomaterial-mediated fibrosis are macrophages, which modulate inflammation, and fibroblasts, which primarily lay down new extracellular matrix. While macrophages and fibroblasts are implicated in driving biomaterial-mediated fibrosis, the signaling pathways and spatiotemporal crosstalk between these cell types remain loosely defined. In this review, the role of M1 and M2 macrophages (and soluble cues) involved in the fibrous encapsulation of biomaterials in vivo is investigated, with additional focus on fibroblast and macrophage crosstalk in vitro along with in vitro models to study the foreign body response. Lastly, several strategies that have been used to specifically modulate macrophage and fibroblast behavior in vitro and in vivo to control biomaterial-mediated fibrosis are highlighted.
Collapse
Affiliation(s)
- Claire E. Witherel
- Drexel University, School of Biomedical Engineering, Science and Health Systems, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, USA
| | - Daniel Abebayehu
- University of Virginia, Department of Biomedical Engineering, School of Engineering & School of Medicine, 415 Lane Road, Charlottesville, Virginia 22904, USA
| | - Thomas H. Barker
- University of Virginia, Department of Biomedical Engineering, School of Engineering & School of Medicine, 415 Lane Road, Charlottesville, Virginia 22904, USA
| | - Kara L. Spiller
- Drexel University, School of Biomedical Engineering, Science and Health Systems, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, USA,
| |
Collapse
|
41
|
Fabrication and Cytocompatibility Evaluation of Psyllium Husk (Isabgol)/Gelatin Composite Scaffolds. Appl Biochem Biotechnol 2019; 188:750-768. [DOI: 10.1007/s12010-019-02958-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022]
|
42
|
Goodarzi P, Aghayan HR, Payab M, Larijani B, Alavi-Moghadam S, Sarvari M, Adibi H, Khatami F, Heravani NF, Hadavandkhani M, Arjmand B. Human Fetal Skin Fibroblast Isolation and Expansion for Clinical Application. Methods Mol Biol 2019; 2109:261-273. [PMID: 31073862 DOI: 10.1007/7651_2019_233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell therapy is one of the most hopeful technologies of regenerative medicine approaches. Among various cells, human skin fibroblasts have been progressively used for wound healing as cell-based therapy purposes. By increasing the age, the number of skin fibroblasts' abilities including cell migration, growth, collagen production, etc. decreases. Hence, use of the fetal source is more beneficent. In this respect, this chapter covers the manufacturing of human fetal skin-derived fibroblasts for clinical application.
Collapse
Affiliation(s)
- Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Adibi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Hadavandkhani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Calejo I, Costa‐Almeida R, Gonçalves AI, Berdecka D, Reis RL, Gomes ME. Bi-directional modulation of cellular interactions in an in vitro co-culture model of tendon-to-bone interface. Cell Prolif 2018; 51:e12493. [PMID: 30105786 PMCID: PMC6528866 DOI: 10.1111/cpr.12493] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES This work aimed at studying in vitro interactions between human tendon-derived cells (hTDCs) and pre-osteoblasts (pre-OBs) that may trigger a cascade of events involved in enthesis regeneration. MATERIALS AND METHODS The effect of 5 osteogenic medium (OM) conditions over the modulation of hTDCs and pre-OBs towards the tenogenic and osteogenic phenotypes, respectively, was studied. Three different medium conditions were chosen for subsequently establishing a direct co-culture system in order to study the expression of bone, tendon and interface-related markers. RESULTS A higher matrix mineralization and ALP activity was observed in co-cultures in the presence of OM. Higher transcription levels of bone- (ALPL, RUNX2, SPP1) and interface-related genes (ACAN, COMP) were found in co-cultures. The expression of aggrecan was influenced by the presence of OM and cell-cell interactions occurring in co-culture. CONCLUSIONS The present work assessed both the influence of OM on cell phenotype modulation and the importance of co-culture models while promoting cell-cell interactions and the exchange of soluble factors in triggering an interface-like phenotype to potentially modulate enthesis regeneration.
Collapse
Affiliation(s)
- I. Calejo
- 3B's Research Group ‐ Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine– Parque de Ciência e TecnologiaBarco, GuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Raquel Costa‐Almeida
- 3B's Research Group ‐ Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine– Parque de Ciência e TecnologiaBarco, GuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Ana Isabel Gonçalves
- 3B's Research Group ‐ Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine– Parque de Ciência e TecnologiaBarco, GuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Dominika Berdecka
- 3B's Research Group ‐ Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine– Parque de Ciência e TecnologiaBarco, GuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Rui Luis Reis
- 3B's Research Group ‐ Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine– Parque de Ciência e TecnologiaBarco, GuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of MinhoBarco, GuimarãesPortugal
| | - Manuela Estima Gomes
- 3B's Research Group ‐ Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine– Parque de Ciência e TecnologiaBarco, GuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of MinhoBarco, GuimarãesPortugal
| |
Collapse
|
44
|
Ahmed MF, El-Sayed AK, Chen H, Zhao R, Jin K, Zuo Q, Zhang Y, Li B. Direct conversion of mouse embryonic fibroblast to osteoblast cells using hLMP-3 with Yamanaka factors. Int J Biochem Cell Biol 2018; 106:84-95. [PMID: 30453092 DOI: 10.1016/j.biocel.2018.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/05/2018] [Accepted: 11/16/2018] [Indexed: 01/14/2023]
Abstract
Large bone defects and bone loss after fractures remain significant challenges for orthopedic surgeons. Our study aims to find an available, applicable and biological treatment for bone regeneration overcoming the limitations in ESC/iPSC technology. We directly reprogrammed the mouse embryonic fibroblast (MEF) into osteoblast cells using different combinations of Yamanaka factors with human lim mineralization protein-3 (hLMP-3). LMP is an intracellular LIM-domain protein acting as an effective positive regulator of the osteoblast differentiation. After transduction, cells were cultured in osteogenic medium, and then examined for osteoblast formation. The expression of osteogenic markers (BMP2, Runx2 and Osterix) during reprogramming and in vitro mineralization assay revealed that the best reprogramming cocktail was (c-Myc - Oct4) with hLMP-3. In addition, both immunofluorescent staining and western blot analysis confirmed that osteocalcin (OCN) expression increased in the cells treated with the c-Myc/Oct4/hLMP3 cocktail than using hLMP-3 alone. Furthermore, this reprogramming cocktail showed efficient healing in an induced femoral bone defect in rat animal model one month after transplantation. In the present study, we reported for the first time the effect of combining Yamanaka factors with hLMP-3 to induce osteoblast cells from MEF both in vitro and in vivo.
Collapse
Affiliation(s)
- Mahmoud F Ahmed
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China; College of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | | | - Hao Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Ruifeng Zhao
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yani Zhang
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
45
|
Differential Proteomic Analysis Predicts Appropriate Applications for the Secretome of Adipose-Derived Mesenchymal Stem/Stromal Cells and Dermal Fibroblasts. Stem Cells Int 2018; 2018:7309031. [PMID: 30158987 PMCID: PMC6109467 DOI: 10.1155/2018/7309031] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/03/2018] [Indexed: 12/12/2022] Open
Abstract
The adult stem cell secretome is currently under investigation as an alternative to cell-based therapy in regenerative medicine, thanks to the remarkable translational opportunity and the advantages in terms of handling and safety. In this perspective, we recently demonstrated the efficient performance of the adipose-derived mesenchymal stem/stromal cell (ASC) secretome in contrasting neuroinflammation in a murine model of diabetic neuropathy, where the administration of factors released by dermal fibroblasts (DFs) did not exert any effect. Up to now, the complex mixture of the constituents of the conditioned medium from ASCs has not been fully deepened, although its appropriate characterization is required in the perspective of a clinical use. Herein, we propose the differential proteomic approach for the identification of the players accounting for the functional effects of the cell secretome with the aim to unravel its appropriate applications. Out of 967 quantified proteins, 34 and 62 factors were found preponderantly or exclusively secreted by ASCs and DFs, respectively. This approach led to the recognition of distinct functions related to the conditioned medium of ASCs and DFs, with the former being involved in the regulation of neuronal death and apoptosis and the latter in bone metabolism and ossification. The proosteogenic effect of DF secretome was validated in vitro on human primary osteoblasts, providing a proof of concept of its osteoinductive potential. Besides discovering new applications of the cell type-specific secretome, the proposed strategy could allow the recognition of the cocktail of bioactive factors which might be responsible for the effects of conditioned media, thus providing a solid rationale to the implementation of a cell-free approach in several clinical scenarios involving tissue regeneration.
Collapse
|