51
|
Fedi A, Vitale C, Giannoni P, Caluori G, Marrella A. Biosensors to Monitor Cell Activity in 3D Hydrogel-Based Tissue Models. SENSORS (BASEL, SWITZERLAND) 2022; 22:1517. [PMID: 35214418 PMCID: PMC8879987 DOI: 10.3390/s22041517] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/06/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
Three-dimensional (3D) culture models have gained relevant interest in tissue engineering and drug discovery owing to their suitability to reproduce in vitro some key aspects of human tissues and to provide predictive information for in vivo tests. In this context, the use of hydrogels as artificial extracellular matrices is of paramount relevance, since they allow closer recapitulation of (patho)physiological features of human tissues. However, most of the analyses aimed at characterizing these models are based on time-consuming and endpoint assays, which can provide only static and limited data on cellular behavior. On the other hand, biosensing systems could be adopted to measure on-line cellular activity, as currently performed in bi-dimensional, i.e., monolayer, cell culture systems; however, their translation and integration within 3D hydrogel-based systems is not straight forward, due to the geometry and materials properties of these advanced cell culturing approaches. Therefore, researchers have adopted different strategies, through the development of biochemical, electrochemical and optical sensors, but challenges still remain in employing these devices. In this review, after examining recent advances in adapting existing biosensors from traditional cell monolayers to polymeric 3D cells cultures, we will focus on novel designs and outcomes of a range of biosensors specifically developed to provide real-time analysis of hydrogel-based cultures.
Collapse
Affiliation(s)
- Arianna Fedi
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunication Engineering (IEIIT), 16149 Genoa, Italy; (A.F.); (C.V.)
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, 16126 Genoa, Italy
| | - Chiara Vitale
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunication Engineering (IEIIT), 16149 Genoa, Italy; (A.F.); (C.V.)
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy;
| | - Paolo Giannoni
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy;
| | - Guido Caluori
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33600 Pessac, France;
- INSERM UMR 1045, Cardiothoracic Research Center of Bordeaux, University of Bordeaux, 33600 Pessac, France
| | - Alessandra Marrella
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunication Engineering (IEIIT), 16149 Genoa, Italy; (A.F.); (C.V.)
| |
Collapse
|
52
|
Cvetkovic C, Patel R, Shetty A, Hogan MK, Anderson M, Basu N, Aghlara-Fotovat S, Ramesh S, Sardar D, Veiseh O, Ward ME, Deneen B, Horner PJ, Krencik R. Assessing Gq-GPCR-induced human astrocyte reactivity using bioengineered neural organoids. J Cell Biol 2022; 221:212997. [PMID: 35139144 PMCID: PMC8842185 DOI: 10.1083/jcb.202107135] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/28/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Astrocyte reactivity can directly modulate nervous system function and immune responses during disease and injury. However, the consequence of human astrocyte reactivity in response to specific contexts and within neural networks is obscure. Here, we devised a straightforward bioengineered neural organoid culture approach entailing transcription factor-driven direct differentiation of neurons and astrocytes from human pluripotent stem cells combined with genetically encoded tools for dual cell-selective activation. This strategy revealed that Gq-GPCR activation via chemogenetics in astrocytes promotes a rise in intracellular calcium followed by induction of immediate early genes and thrombospondin 1. However, astrocytes also undergo NF-κB nuclear translocation and secretion of inflammatory proteins, correlating with a decreased evoked firing rate of cocultured optogenetic neurons in suboptimal conditions, without overt neurotoxicity. Altogether, this study clarifies the intrinsic reactivity of human astrocytes in response to targeting GPCRs and delivers a bioengineered approach for organoid-based disease modeling and preclinical drug testing.
Collapse
Affiliation(s)
- Caroline Cvetkovic
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | | | - Arya Shetty
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | - Matthew K Hogan
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | - Morgan Anderson
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | - Nupur Basu
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | - Samira Aghlara-Fotovat
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX.,Department of Bioengineering, Rice University, Houston, TX
| | | | | | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | | | - Philip J Horner
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | - Robert Krencik
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| |
Collapse
|
53
|
Decellularized Alstroemeria flower stem modified with chitosan for tissue engineering purposes: A cellulose/chitosan scaffold. Int J Biol Macromol 2022; 204:321-332. [PMID: 35149092 DOI: 10.1016/j.ijbiomac.2022.02.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/18/2022]
Abstract
Utilizing plant-based scaffolds has pulled in the consideration of tissue engineers. Plant tissues own different structures with particular porosity and structure. In this study, the stem of the Alstroemeria flower was designated for decellularization to fabricate a new scaffold. The stems were decellularized and called AFSP and then modified by chitosan and named AFSPC. Osteoblast precursor cell line was employed to assess the biological potential of the final scaffolds. The results uncovered that AFSP owns linear microchannels with a smooth surface. AFSPC delineated uniform chitosan coating on the walls with appropriate roughness. AFSPC showed higher potential in swelling, degradation, diffusion, and having a porous structure than AFSP. Modification with chitosan improved mechanical behavior. Biological assays depicted no cytotoxicity for AFSP and AFSPC. AFSPC showed good cell attachment, proliferation, and migration. In conclusion, modified tissue plants can be a good candidate for tissue engineering of both soft and hard tissues.
Collapse
|
54
|
Zargarzadeh M, Silva AS, Nunes C, Coimbra MA, Custódio CA, Mano JF. Self-glucose feeding hydrogels by enzyme empowered degradation for 3D cell culture. MATERIALS HORIZONS 2022; 9:694-707. [PMID: 34825909 DOI: 10.1039/d0mh01982h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Hydrogels have been used in combination with cells for several biomedical and biotechnological applications. Nevertheless, the use of bulk hydrogels has exhibited severe limitations in diffusion of oxygen, nutrients, and metabolites. Here, a support for cell culture is reported where glucose is generated in situ by the own hydrogel degradation, allowing cell survival and function while promoting tissue growth. For this purpose, laminaran (or laminarin)-based hydrogels were fabricated, immobilizing the adequate enzymes to obtain structural platforms for 3D cell culture and providing glucose feeding for metabolic activity of cells through polysaccharide degradation. We demonstrate that tumor A549 cells and human mesenchymal stem cells (hMSCs) can use the glucose resultant from the hydrogel degradation to survive and grow in non-added glucose cell culture medium. Additionally, in vivo biocompatibility and biodegradability of laminaran-based hydrogels were explored for the first time. The self-feeding hydrogels exhibited high potential in cell survival compared to native cell-laden laminaran hydrogels over two weeks of sub-cutaneous implantation. Such bioscaffolds with enzyme-empowered degradation capacity can be applied in diverse biotechnological contexts such as tissue regeneration devices, biofactories, disease models, and cell delivery systems.
Collapse
Affiliation(s)
- Mehrzad Zargarzadeh
- CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal.
| | - A Sofia Silva
- CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal.
| | - Cláudia Nunes
- CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal.
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Manuel A Coimbra
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Catarina A Custódio
- CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal.
| | - João F Mano
- CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal.
| |
Collapse
|
55
|
Lipreri MV, Baldini N, Graziani G, Avnet S. Perfused Platforms to Mimic Bone Microenvironment at the Macro/Milli/Microscale: Pros and Cons. Front Cell Dev Biol 2022; 9:760667. [PMID: 35047495 PMCID: PMC8762164 DOI: 10.3389/fcell.2021.760667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
As life expectancy increases, the population experiences progressive ageing. Ageing, in turn, is connected to an increase in bone-related diseases (i.e., osteoporosis and increased risk of fractures). Hence, the search for new approaches to study the occurrence of bone-related diseases and to develop new drugs for their prevention and treatment becomes more pressing. However, to date, a reliable in vitro model that can fully recapitulate the characteristics of bone tissue, either in physiological or altered conditions, is not available. Indeed, current methods for modelling normal and pathological bone are poor predictors of treatment outcomes in humans, as they fail to mimic the in vivo cellular microenvironment and tissue complexity. Bone, in fact, is a dynamic network including differently specialized cells and the extracellular matrix, constantly subjected to external and internal stimuli. To this regard, perfused vascularized models are a novel field of investigation that can offer a new technological approach to overcome the limitations of traditional cell culture methods. It allows the combination of perfusion, mechanical and biochemical stimuli, biological cues, biomaterials (mimicking the extracellular matrix of bone), and multiple cell types. This review will discuss macro, milli, and microscale perfused devices designed to model bone structure and microenvironment, focusing on the role of perfusion and encompassing different degrees of complexity. These devices are a very first, though promising, step for the development of 3D in vitro platforms for preclinical screening of novel anabolic or anti-catabolic therapeutic approaches to improve bone health.
Collapse
Affiliation(s)
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Biomedical Science and Technologies Lab, IRCSS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gabriela Graziani
- Laboratory for NanoBiotechnology (NaBi), IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
56
|
Alginate Hydrogel Microtubes for Salivary Gland Cell Organization and Cavitation. Bioengineering (Basel) 2022; 9:bioengineering9010038. [PMID: 35049747 PMCID: PMC8773299 DOI: 10.3390/bioengineering9010038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/25/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022] Open
Abstract
Understanding the different regulatory functions of epithelial and mesenchymal cell types in salivary gland development and cellular organization is essential for proper organoid formation and salivary gland tissue regeneration. Here, we demonstrate a biocompatible platform using pre-formed alginate hydrogel microtubes to facilitate direct epithelial–mesenchymal cell interaction for 3D salivary gland cell organization, which allows for monitoring cellular organization while providing a protective barrier from cell-cluster loss during medium changes. Using mouse salivary gland ductal epithelial SIMS cells as the epithelial model cell type and NIH 3T3 fibroblasts or primary E16 salivary mesenchyme cells as the stromal model cell types, self-organization from epithelial–mesenchymal interaction was examined. We observed that epithelial and mesenchymal cells undergo aggregation on day 1, cavitation by day 4, and generation of an EpCAM-expressing epithelial cell layer as early as day 7 of the co-culture in hydrogel microtubes, demonstrating the utility of hydrogel microtubes to facilitate heterotypic cell–cell interactions to form cavitated organoids. Thus, pre-formed alginate microtubes are a promising co-culture method for further understanding epithelial and mesenchymal interaction during tissue morphogenesis and for future practical applications in regenerative medicine.
Collapse
|
57
|
Wang J, Lin Y, Chen X, Liu Y, Zhou T. Mesenchymal stem cells: A new therapeutic tool for chronic kidney disease. Front Cell Dev Biol 2022; 10:910592. [PMID: 36268508 PMCID: PMC9577598 DOI: 10.3389/fcell.2022.910592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/12/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic kidney disease (CKD) has a major impact on public health, which could progress to end-stage kidney disease (ESRD) and consume many medical resources. Currently, the treatment for CKD has many flaws, so more effective treatment tools are urgently required for CKD. Mesenchymal stem cells (MSCs) are primitive cells with self-renewal and proliferation capacity and differentiation potential. Extensive preclinical and clinical data has shown that cell-based therapies using MSCs can modulate immunity, inhibit inflammatory factors, and improve renal function in CKD, suggesting that MSCs have the potential to be a new, effective therapeutic tool for CKD. In this review, we will describe different kinds of MSCs and MSCs products for the treatment of CKD in experimental models and clinical trials, potential signaling pathways, therapeutic efficacy, and critical issues that need to be addressed before therapeutic application in humans.
Collapse
Affiliation(s)
- Jiali Wang
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yongda Lin
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xiutian Chen
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yiping Liu
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| |
Collapse
|
58
|
Upenieks A, Montgomery-Song A, Santerre JP, Kandel RA. Development of a Perfusion Reactor for Intervertebral Disk Regeneration. Tissue Eng Part C Methods 2022; 28:12-22. [PMID: 35018812 DOI: 10.1089/ten.tec.2021.0216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A tissue-engineered biological disk replacement has been proposed as a promising approach for the treatment of degenerative disk disease. A perfusion bioreactor would be a logical consideration to facilitate this scale-up as such reactors have been shown to improve nutrient delivery and provide beneficial mechanical forces that support the cultivation of large three-dimensional constructs. It was hypothesized that perfusion culture of tissue-engineered intervertebral disk (IVD) tissues would be capable of generating outer annulus fibrosus (oAF) and nucleus pulposus (NP) tissues comparable with established spinner reactor or static cultures, respectively, without compromising cellular viability, nutrient delivery, and tissue formation. In this study, the perfusion grown oAF and NP tissues did not show a significant difference in extracellular matrix (ECM) quantity or cellular phenotype when compared with their control conditions. In addition, they maintained cellular viability at the center core of the tissues and received enhanced diffusion of medium throughout the tissue when compared with static conditions. This study lays the groundwork for future studies to grow an entire IVD tissue to a physiologically relevant size.
Collapse
Affiliation(s)
- Alexander Upenieks
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada
| | - Aaryn Montgomery-Song
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, and Toronto, Ontario, Canada
| | - John Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada.,Translational Biology and Engineering Program and Faculty of Dentistry, Toronto, Ontario, Canada
| | - Rita A Kandel
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, and Toronto, Ontario, Canada
| |
Collapse
|
59
|
Thorp H, Kim K, Bou-Ghannam S, Kondo M, Maak T, Grainger DW, Okano T. Enhancing chondrogenic potential via mesenchymal stem cell sheet multilayering. Regen Ther 2021; 18:487-496. [PMID: 34926734 PMCID: PMC8645782 DOI: 10.1016/j.reth.2021.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
Advanced tissue engineering approaches for direct articular cartilage replacement in vivo employ mesenchymal stem cell (MSC) sources, exploiting innate chondrogenic potential to fabricate hyaline-like constructs in vitro within three-dimensional (3D) culture conditions. Cell sheet technology represents one such advanced 3D scaffold-free cell culture platform, and previous work has shown that 3D MSC sheets are capable of in vitro hyaline-like chondrogenic differentiation. The present study aims to build upon this understanding and elucidate the effects of an established cell sheet manipulation technique, cell sheet multilayering, on fabrication of MSC-derived hyaline-like cartilage 3D layered constructs in vitro. To achieve this goal, multilayered MSC sheets are prepared and assessed for structural and biochemical transitions throughout chondrogenesis. Results support MSC multilayering as a means of increasing construct thickness and 3D cellular interactions related to in vitro chondrogenesis, including N-cadherin, connexin 43, and integrin β-1. Data indicate that increasing construct thickness from 14 μm (1-layer construct) to 25 μm (2-layer construct) increases these cellular interactions and subsequent in vitro MSC chondrogenesis. However, a clear initial thickness threshold (33 μm - 3-layer construct) is evident that decreases the rate and extent of in vitro chondrogenesis, specifically chondrogenic gene expressions (Sox9, aggrecan, type II collagen) and sulfated proteoglycan accumulation in deposited extracellular matrix (ECM). Together, these data support the utility of cell sheet multilayering as a platform for tailoring construct thickness and subsequent MSC chondrogenesis for future articular cartilage regeneration applications.
Collapse
Affiliation(s)
- Hallie Thorp
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Sophia Bou-Ghannam
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Makoto Kondo
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Travis Maak
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, UT, USA
| | - David W. Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, Japan
| |
Collapse
|
60
|
Anand R, Nimi N, Sivadas VP, Merlin Rajesh Lal LP, Nair PD. Dual crosslinked pullulan-gelatin cryogel scaffold for chondrocyte-mediated cartilage repair: synthesis, characterization and in vitroevaluation. Biomed Mater 2021; 17. [PMID: 34700303 DOI: 10.1088/1748-605x/ac338b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/26/2021] [Indexed: 11/11/2022]
Abstract
Cryogels, a subset of hydrogels, have recently drawn attention for cartilage tissue engineering due to its inherent microporous architecture and good mechanical properties. In this study a dual crosslinked pullulan-gelatin cryogel (PDAG) scaffold was synthesized by crosslinking gelatin with oxidized pullulan by Schiff's base reaction followed by cryogelation. Chondrocytes seeded within the PDAG scaffolds and cultured for 21 din vitrodemonstrated enhanced cell proliferation, enhanced production of cartilage-specific extracellular matrix and up-regulated sulfated glycosaminoglycan without altering the articular chondrocyte phenotype. Quantitative reverse transcription-polymerase chain reaction-based gene expression studies, immunofluorescence, and histological studies demonstrated that the PDAG scaffold significantly enhanced the expression of chondrogenic marker genes such as type II collagen, aggrecan, and SOX9. Taken together, these results demonstrated that PDAG scaffold prepared by sequential Schiff's base reaction and cryogelation would be a promising cell-responsive scaffold for cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Resmi Anand
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India.,Inter University Centre for Biomedical Research and Super Speciality Hospital, Kottayam, Kerala 686009, India
| | - N Nimi
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - V P Sivadas
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - L P Merlin Rajesh Lal
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - Prabha D Nair
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| |
Collapse
|
61
|
Wang WY, Kent RN, Huang SA, Jarman EH, Shikanov EH, Davidson CD, Hiraki HL, Lin D, Wall MA, Matera DL, Shin JW, Polacheck WJ, Shikanov A, Baker BM. Direct comparison of angiogenesis in natural and synthetic biomaterials reveals that matrix porosity regulates endothelial cell invasion speed and sprout diameter. Acta Biomater 2021; 135:260-273. [PMID: 34469789 PMCID: PMC8595798 DOI: 10.1016/j.actbio.2021.08.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022]
Abstract
Vascularization of large, diffusion-hindered biomaterial implants requires an understanding of how extracellular matrix (ECM) properties regulate angiogenesis. Sundry biomaterials assessed across many disparate angiogenesis assays have highlighted ECM determinants that influence this complex multicellular process. However, the abundance of material platforms, each with unique parameters to model endothelial cell (EC) sprouting presents additional challenges of interpretation and comparison between studies. In this work we directly compared the angiogenic potential of commonly utilized natural (collagen and fibrin) and synthetic dextran vinyl sulfone (DexVS) hydrogels in a multiplexed angiogenesis-on-a-chip platform. Modulating matrix density of collagen and fibrin hydrogels confirmed prior findings that increases in matrix density correspond to increased EC invasion as connected, multicellular sprouts, but with decreased invasion speeds. Angiogenesis in synthetic DexVS hydrogels, however, resulted in fewer multicellular sprouts. Characterizing hydrogel Young's modulus and permeability (a measure of matrix porosity), we identified matrix permeability to significantly correlate with EC invasion depth and sprout diameter. Although microporous collagen and fibrin hydrogels produced lumenized sprouts in vitro, they rapidly resorbed post-implantation into the murine epididymal fat pad. In contrast, DexVS hydrogels proved comparatively stable. To enhance angiogenesis within DexVS hydrogels, we incorporated sacrificial microgels to generate cell-scale pores throughout the hydrogel. Microporous DexVS hydrogels resulted in lumenized sprouts in vitro and enhanced cell invasion in vivo. Towards the design of vascularized biomaterials for long-term regenerative therapies, this work suggests that synthetic biomaterials offer improved size and shape control following implantation and that tuning matrix porosity may better support host angiogenesis. STATEMENT OF SIGNIFICANCE: Understanding how extracellular matrix properties govern angiogenesis will inform biomaterial design for engineering vascularized implantable grafts. Here, we utilized a multiplexed angiogenesis-on-a-chip platform to compare the angiogenic potential of natural (collagen and fibrin) and synthetic dextran vinyl sulfone (DexVS) hydrogels. Characterization of matrix properties and sprout morphometrics across these materials points to matrix porosity as a critical regulator of sprout invasion speed and diameter, supported by the observation that nanoporous DexVS hydrogels yielded endothelial cell sprouts that were not perfusable. To enhance angiogenesis into synthetic hydrogels, we incorporated sacrificial microgels to generate microporosity. We find that microporosity increased sprout diameter in vitro and cell invasion in vivo. This work establishes a composite materials approach to enhance the vascularization of synthetic hydrogels.
Collapse
Affiliation(s)
- William Y Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Stephanie A Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27514, United States
| | - Evan H Jarman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Eve H Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Christopher D Davidson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Harrison L Hiraki
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Daphne Lin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Monica A Wall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Daniel L Matera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine & Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, United States
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27514, United States; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, United States
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Macromolecular Science & Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
62
|
Farzin A, Hassan S, Teixeira LSM, Gurian M, Crispim JF, Manhas V, Carlier A, Bae H, Geris L, Noshadi I, Shin SR, Leijten J. Self-Oxygenation of Tissues Orchestrates Full-Thickness Vascularization of Living Implants. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2100850. [PMID: 34924912 PMCID: PMC8680410 DOI: 10.1002/adfm.202100850] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Indexed: 05/13/2023]
Abstract
Bioengineering of tissues and organs has the potential to generate functional replacement organs. However, achieving the full-thickness vascularization that is required for long-term survival of living implants has remained a grand challenge, especially for clinically sized implants. During the pre-vascular phase, implanted engineered tissues are forced to metabolically rely on the diffusion of nutrients from adjacent host-tissue, which for larger living implants results in anoxia, cell death, and ultimately implant failure. Here it is reported that this challenge can be addressed by engineering self-oxygenating tissues, which is achieved via the incorporation of hydrophobic oxygen-generating micromaterials into engineered tissues. Self-oxygenation of tissues transforms anoxic stresses into hypoxic stimulation in a homogenous and tissue size-independent manner. The in situ elevation of oxygen tension enables the sustained production of high quantities of angiogenic factors by implanted cells, which are offered a metabolically protected pro-angiogenic microenvironment. Numerical simulations predict that self-oxygenation of living tissues will effectively orchestrate rapid full-thickness vascularization of implanted tissues, which is empirically confirmed via in vivo experimentation. Self-oxygenation of tissues thus represents a novel, effective, and widely applicable strategy to enable the vascularization living implants, which is expected to advance organ transplantation and regenerative medicine applications.
Collapse
Affiliation(s)
- Ali Farzin
- Division of Engineering in Medicine Department of Medicine Brigham and Women's Hospital Harvard Medical School Cambridge, MA 02139, USA
| | - Shabir Hassan
- Division of Engineering in Medicine Department of Medicine Brigham and Women's Hospital Harvard Medical School Cambridge, MA 02139, USA
| | - Liliana S Moreira Teixeira
- Department of Developmental BioEngineering Technical Medical Centre University of Twente Enschede, The Netherlands
| | - Melvin Gurian
- Department of Developmental BioEngineering Technical Medical Centre University of Twente Enschede, The Netherlands
| | - João F Crispim
- Department of Developmental BioEngineering Technical Medical CentreUniversity of Twente Enschede, The Netherlands
| | - Varun Manhas
- Biomechanics Research Unit GIGA In Silico Medicine University of Liège Chemin des Chevreuils 1, B52/3, Liège 4000, Belgium
| | - Aurélie Carlier
- Laboratory for Cell Biology-Inspired Tissue Engineering MERLN Institute University of Maastricht Maastricht, The Netherlands
| | - Hojae Bae
- KU Convergence Science and Technology Institute Department of Stem Cell and Regenerative Biotechnology Konkuk University Seoul 05029, Republic of Korea
| | - Liesbet Geris
- Biomechanics Research Unit GIGA In Silico Medicine University of Liège Chemin des Chevreuils 1, B52/3, Liège 4000, Belgium
| | - Iman Noshadi
- Department of Bioengineering University of California Riverside, CA 92521, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine Department of Medicine Brigham and Women's Hospital Harvard Medical School Cambridge, MA 02139, USA
| | - Jeroen Leijten
- Division of Engineering in Medicine Department of Medicine Brigham and Women's Hospital Harvard Medical School Cambridge, MA 02139, USA
| |
Collapse
|
63
|
Paradiso F, Serpelloni S, Francis LW, Taraballi F. Mechanical Studies of the Third Dimension in Cancer: From 2D to 3D Model. Int J Mol Sci 2021; 22:10098. [PMID: 34576261 PMCID: PMC8472581 DOI: 10.3390/ijms221810098] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
From the development of self-aggregating, scaffold-free multicellular spheroids to the inclusion of scaffold systems, 3D models have progressively increased in complexity to better mimic native tissues. The inclusion of a third dimension in cancer models allows researchers to zoom out from a significant but limited cancer cell research approach to a wider investigation of the tumor microenvironment. This model can include multiple cell types and many elements from the extracellular matrix (ECM), which provides mechanical support for the tissue, mediates cell-microenvironment interactions, and plays a key role in cancer cell invasion. Both biochemical and biophysical signals from the extracellular space strongly influence cell fate, the epigenetic landscape, and gene expression. Specifically, a detailed mechanistic understanding of tumor cell-ECM interactions, especially during cancer invasion, is lacking. In this review, we focus on the latest achievements in the study of ECM biomechanics and mechanosensing in cancer on 3D scaffold-based and scaffold-free models, focusing on each platform's level of complexity, up-to-date mechanical tests performed, limitations, and potential for further improvements.
Collapse
Affiliation(s)
- Francesca Paradiso
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Stefano Serpelloni
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| | - Lewis W. Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| |
Collapse
|
64
|
Kim KS, Joo HJ, Choi SC, Kim JH, Park CY, Song MH, Noh JM, Cha JJ, Hong SJ, Ahn TH, Kim MN, Na JE, Rhyu IJ, Lim DS. Transplantation of 3D bio-printed cardiac mesh improves cardiac function and vessel formation via ANGPT1/Tie2 pathway in rats with acute myocardial infarction. Biofabrication 2021; 13. [PMID: 34404035 DOI: 10.1088/1758-5090/ac1e78] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/17/2021] [Indexed: 12/31/2022]
Abstract
A novel tissue engineering strategy using 3D bio-print technology has become a promising therapeutic method for acute myocardial infarction (AMI) in an animal model. However, the application of 3D bio-printed tissue remains limited due to poor graft survival. Therefore, it is a scientific priority to enhance graft survival by precisely adjusting the 3D environment of encapsulated cells. In this study, novel transplantable 3D cardiac mesh (cMesh) tissue with a porous mesh structure was presented using human cardiomyocytes, human cardiac fibroblasts, and gelatin-methacryloyl-collagen hydrogel. Cardiomyocytes and cardiac fibroblasts were well spreaded. The cardiomyocytes were connected with a gap junction channel in bio-printed cMesh and a 3D cardiac patch with an aggregated structure. Porous cMesh demonstrated structural advantages by increased phosphorylation of mTOR, AKT, and ERK signals associated with cell survival. Transplanted cMesh in rats with AMI improved long-term graft survival, vessel formation, and stabilization, reduced fibrosis, increased left ventricle thickness, and enhanced cardiac function. Our results suggest that porous cMesh provides structural advantages and a positive therapeutic effect in an AMI animal model.
Collapse
Affiliation(s)
- Kyung Seob Kim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Seung-Cheol Choi
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jong-Ho Kim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Chi-Yeon Park
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Myeong-Hwa Song
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Ji-Min Noh
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jung-Joon Cha
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Soon Jun Hong
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Tae Hoon Ahn
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Mi-Na Kim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Ji Eun Na
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
65
|
Rahimnejad M, Nasrollahi Boroujeni N, Jahangiri S, Rabiee N, Rabiee M, Makvandi P, Akhavan O, Varma RS. Prevascularized Micro-/Nano-Sized Spheroid/Bead Aggregates for Vascular Tissue Engineering. NANO-MICRO LETTERS 2021; 13:182. [PMID: 34409511 PMCID: PMC8374027 DOI: 10.1007/s40820-021-00697-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/13/2021] [Indexed: 05/02/2023]
Abstract
Efficient strategies to promote microvascularization in vascular tissue engineering, a central priority in regenerative medicine, are still scarce; nano- and micro-sized aggregates and spheres or beads harboring primitive microvascular beds are promising methods in vascular tissue engineering. Capillaries are the smallest type and in numerous blood vessels, which are distributed densely in cardiovascular system. To mimic this microvascular network, specific cell components and proangiogenic factors are required. Herein, advanced biofabrication methods in microvascular engineering, including extrusion-based and droplet-based bioprinting, Kenzan, and biogripper approaches, are deliberated with emphasis on the newest works in prevascular nano- and micro-sized aggregates and microspheres/microbeads.
Collapse
Affiliation(s)
- Maedeh Rahimnejad
- Biomedical Engineering Institute, School of Medicine, Université de Montréal, Montreal, Canada
- Research Centre, Centre Hospitalier de L'Université de Montréal (CRCHUM), Montreal, Canada
| | | | - Sepideh Jahangiri
- Research Centre, Centre Hospitalier de L'Université de Montréal (CRCHUM), Montreal, Canada
- Department of Biomedical Sciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Navid Rabiee
- Department of Physics, Sharif University of Technology, P.O. Box 11155-9161, Tehran, Iran.
| | - Mohammad Rabiee
- Biomaterial Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interfaces, Istituto Italiano Di Tecnologia, viale Rinaldo Piaggio 34, 56 025, Pontedera, Pisa, Italy
| | - Omid Akhavan
- Department of Physics, Sharif University of Technology, P.O. Box 11155-9161, Tehran, Iran.
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
66
|
Petry F, Salzig D. Impact of Bioreactor Geometry on Mesenchymal Stem Cell Production in Stirred‐Tank Bioreactors. CHEM-ING-TECH 2021. [DOI: 10.1002/cite.202100041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Florian Petry
- University of Applied Sciences Mittelhessen Institute of Bioprocess Engineering and Pharmaceutical Technology Wiesenstraße 14 35390 Giessen Germany
| | - Denise Salzig
- University of Applied Sciences Mittelhessen Institute of Bioprocess Engineering and Pharmaceutical Technology Wiesenstraße 14 35390 Giessen Germany
| |
Collapse
|
67
|
Naumova N, Iop L. Bioengineering the Cardiac Conduction System: Advances in Cellular, Gene, and Tissue Engineering for Heart Rhythm Regeneration. Front Bioeng Biotechnol 2021; 9:673477. [PMID: 34409019 PMCID: PMC8365186 DOI: 10.3389/fbioe.2021.673477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/24/2021] [Indexed: 01/01/2023] Open
Abstract
Heart rhythm disturbances caused by different etiologies may affect pediatric and adult patients with life-threatening consequences. When pharmacological therapy is ineffective in treating the disturbances, the implantation of electronic devices to control and/or restore normal heart pacing is a unique clinical management option. Although these artificial devices are life-saving, they display many limitations; not least, they do not have any capability to adapt to somatic growth or respond to neuroautonomic physiological changes. A biological pacemaker could offer a new clinical solution for restoring heart rhythms in the conditions of disorder in the cardiac conduction system. Several experimental approaches, such as cell-based, gene-based approaches, and the combination of both, for the generation of biological pacemakers are currently established and widely studied. Pacemaker bioengineering is also emerging as a technology to regenerate nodal tissues. This review analyzes and summarizes the strategies applied so far for the development of biological pacemakers, and discusses current translational challenges toward the first-in-human clinical application.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
68
|
Lehder EF, Ashcroft IA, Wildman RD, Ruiz-Cantu LA, Maskery I. A multiscale optimisation method for bone growth scaffolds based on triply periodic minimal surfaces. Biomech Model Mechanobiol 2021; 20:2085-2096. [PMID: 34318358 PMCID: PMC8595174 DOI: 10.1007/s10237-021-01496-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/10/2021] [Indexed: 11/09/2022]
Abstract
Tissue engineered bone scaffolds are potential alternatives to bone allografts and autografts. Porous scaffolds based on triply periodic minimal surfaces (TPMS) are good candidates for tissue growth because they offer high surface-to-volume ratio, have tailorable stiffness, and can be easily fabricated by additive manufacturing. However, the range of TPMS scaffold types is extensive, and it is not yet clear which type provides the fastest cell or tissue growth while being sufficiently stiff to act as a bone graft. Nor is there currently an established methodology for TPMS bone scaffold design which can be quickly adopted by medical designers or biologists designing implants. In this study, we examine six TPMS scaffold types for use as tissue growth scaffolds and propose a general methodology to optimise their geometry. At the macro-scale, the optimisation routine ensures a scaffold stiffness within suitable limits for bone, while at the micro-scale it maximises the cell growth rate. The optimisation procedure also ensures the scaffold pores are of sufficient diameter to allow oxygen and nutrient delivery via capillaries. Of the examined TPMS structures, the Lidinoid and Split P cell types provide the greatest cell growth rates and are therefore the best candidates for bone scaffolds.
Collapse
Affiliation(s)
- E F Lehder
- Centre for Additive Manufacturing, Faculty of Engineering, The University of Nottingham, Nottingham, NG7 2RD, UK
| | - I A Ashcroft
- Centre for Additive Manufacturing, Faculty of Engineering, The University of Nottingham, Nottingham, NG7 2RD, UK
| | - R D Wildman
- Centre for Additive Manufacturing, Faculty of Engineering, The University of Nottingham, Nottingham, NG7 2RD, UK
| | - L A Ruiz-Cantu
- Centre for Additive Manufacturing, Faculty of Engineering, The University of Nottingham, Nottingham, NG7 2RD, UK
| | - I Maskery
- Centre for Additive Manufacturing, Faculty of Engineering, The University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
69
|
Mirani B, Stefanek E, Godau B, Hossein Dabiri SM, Akbari M. Microfluidic 3D Printing of a Photo-Cross-Linkable Bioink Using Insights from Computational Modeling. ACS Biomater Sci Eng 2021; 7:3269-3280. [PMID: 34142796 DOI: 10.1021/acsbiomaterials.1c00084] [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: 11/29/2022]
Abstract
Three-dimensional (3D) bioprinting of photo-cross-linkable hydrogel precursors has attracted great interest in various tissue engineering and drug screening applications, as the biochemical and biophysical properties of the resultant hydrogel structures can be tuned spatiotemporally to provide cells with physiologically relevant microenvironments. In particular, these bioinks benefit from great biofunctional versatility that can be designed to direct cells toward a desired behavior. Despite significant progress in the field, the 3D printing of cell-laden photo-cross-linkable bioinks with low polymer concentrations has remained a challenge, as rapidly stabilizing these bioinks and transforming them to hydrogel filaments is hindered by their low viscosity. Additionally, reaching an optimized print condition has often been challenging due to the large number of print parameters involved in 3D bioprinting setups. Therefore, computational modeling has occasionally been employed to understand the impact of various print parameters and reduce the time and resources required to determine these effects in experimental settings. Here, we report a novel 3D bioprinting strategy for fabricating hydrogel fibrous structures of gelatin methacryloyl (GelMA) with superior control over polymer concentration, particularly in a relatively low range from ∼1% (w/v) to 6% (w/v), using a microfluidic printhead. The printhead features a coaxial core-sheath flow, coupled with a photo-cross-linking system, allowing for the in situ cross-linking of GelMA and the generation of hydrogel filaments. A computational model was developed to determine the optimal ranges of process parameters and inform about the diffusive and fluid dynamic behavior of the coaxial flow. The cytocompatibility of the biofabrication system was determined via bioprinting cell-laden bioinks containing U87-MG cells. Notably, the established pipeline from computational modeling to bioprinting has great potential to be applied to a wide range of photo-cross-linkable bioinks to generate living tissues with various material and cellular characteristics.
Collapse
Affiliation(s)
- Bahram Mirani
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Biomedical Research, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Evan Stefanek
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Brent Godau
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Biomedical Research, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Seyed Mohammad Hossein Dabiri
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Biomedical Research, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Centre for Biomedical Research, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.,Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| |
Collapse
|
70
|
Hughes DL, Hughes A, Soonawalla Z, Mukherjee S, O’Neill E. Dynamic Physiological Culture of Ex Vivo Human Tissue: A Systematic Review. Cancers (Basel) 2021; 13:2870. [PMID: 34201273 PMCID: PMC8229413 DOI: 10.3390/cancers13122870] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022] Open
Abstract
Conventional static culture fails to replicate the physiological conditions that exist in vivo. Recent advances in biomedical engineering have resulted in the creation of novel dynamic culturing systems that permit the recapitulation of normal physiological processes ex vivo. Whilst the physiological benefit for its use in the culture of two-dimensional cellular monolayer has been validated, its role in the context of primary human tissue culture has yet to be determined. This systematic review identified 22 articles that combined dynamic physiological culture techniques with primary human tissue culture. The most frequent method described (55%) utilised dynamic perfusion culture. A diverse range of primary human tissue was successfully cultured. The median duration of successful ex vivo culture of primary human tissue for all articles was eight days; however, a wide range was noted (5 h-60 days). Six articles (27%) reported successful culture of primary human tissue for greater than 20 days. This review illustrates the physiological benefit of combining dynamic culture with primary human tissue culture in both long-term culture success rates and preservation of native functionality of the tissue ex vivo. Further research efforts should focus on developing precise biochemical sensors that would allow for real-time monitoring and automated self-regulation of the culture system in order to maintain homeostasis. Combining these techniques allows the creation of an accurate system that can be used to gain a greater understanding of human physiology.
Collapse
Affiliation(s)
- Daniel Ll Hughes
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (D.L.H.); (S.M.)
| | - Aron Hughes
- Undergraduate Centre, Cardiff University Medical School, Cardiff CF14 4YS, UK;
| | - Zahir Soonawalla
- Department of Hepatobiliary and Pancreatic Surgery, Oxford University Hospitals NHS, Oxford OX3 7LE, UK;
| | - Somnath Mukherjee
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (D.L.H.); (S.M.)
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (D.L.H.); (S.M.)
| |
Collapse
|
71
|
Nazeer MA, Karaoglu IC, Ozer O, Albayrak C, Kizilel S. Neovascularization of engineered tissues for clinical translation: Where we are, where we should be? APL Bioeng 2021; 5:021503. [PMID: 33834155 PMCID: PMC8024034 DOI: 10.1063/5.0044027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
One of the key challenges in engineering three-dimensional tissue constructs is the development of a mature microvascular network capable of supplying sufficient oxygen and nutrients to the tissue. Recent angiogenic therapeutic strategies have focused on vascularization of the constructed tissue, and its integration in vitro; these strategies typically combine regenerative cells, growth factors (GFs) with custom-designed biomaterials. However, the field needs to progress in the clinical translation of tissue engineering strategies. The article first presents a detailed description of the steps in neovascularization and the roles of extracellular matrix elements such as GFs in angiogenesis. It then delves into decellularization, cell, and GF-based strategies employed thus far for therapeutic angiogenesis, with a particularly detailed examination of different methods by which GFs are delivered in biomaterial scaffolds. Finally, interdisciplinary approaches involving advancement in biomaterials science and current state of technological development in fabrication techniques are critically evaluated, and a list of remaining challenges is presented that need to be solved for successful translation to the clinics.
Collapse
Affiliation(s)
| | | | - Onur Ozer
- Biomedical Sciences and Engineering, Koç University, Istanbul 34450, Turkey
| | - Cem Albayrak
- Authors to whom correspondence should be addressed: and
| | - Seda Kizilel
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
72
|
Mansouri M, Leipzig ND. Advances in removing mass transport limitations for more physiologically relevant in vitro 3D cell constructs. BIOPHYSICS REVIEWS 2021; 2:021305. [PMID: 38505119 PMCID: PMC10903443 DOI: 10.1063/5.0048837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/31/2021] [Indexed: 03/21/2024]
Abstract
Spheroids and organoids are promising models for biomedical applications ranging from human disease modeling to drug discovery. A main goal of these 3D cell-based platforms is to recapitulate important physiological parameters of their in vivo organ counterparts. One way to achieve improved biomimetic architectures and functions is to culture cells at higher density and larger total numbers. However, poor nutrient and waste transport lead to low stability, survival, and functionality over extended periods of time, presenting outstanding challenges in this field. Fortunately, important improvements in culture strategies have enhanced the survival and function of cells within engineered microtissues/organs. Here, we first discuss the challenges of growing large spheroids/organoids with a focus on mass transport limitations, then highlight recent tools and methodologies that are available for producing and sustaining functional 3D in vitro models. This information points toward the fact that there is a critical need for the continued development of novel cell culture strategies that address mass transport in a physiologically relevant human setting to generate long-lasting and large-sized spheroids/organoids.
Collapse
Affiliation(s)
- Mona Mansouri
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| | - Nic D. Leipzig
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
73
|
Damaraju SM, Mintz BR, Park JG, Gandhi A, Saini S, Molnar JA. Skin substitutes with noncultured autologous skin cell suspension heal porcine full-thickness wounds in a one-stage procedure. Int Wound J 2021; 19:188-201. [PMID: 34036743 PMCID: PMC8684857 DOI: 10.1111/iwj.13615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 11/28/2022] Open
Abstract
Clinical application of skin substitute is typically a two-stage procedure with application of skin substitute matrix to the wound followed by engraftment of a split-thickness skin graft (STSG). This two-stage procedure requires multiple interventions, increasing the time until the wound is epithelialised. In this study, the feasibility of a one-stage procedure by combining bioengineered collagen-chondroitin-6-sulfate (DS1) or decellularised fetal bovine skin substitute (DS2) with autologous skin cell suspension (ASCS) in a porcine full-thickness wound healing model was evaluated. Twelve full-thickness excisional wounds on the backs of pigs received one of six different treatments: empty; ASCS; DS1 with or without ASCS; DS2 with or without ASCS. The ASCS was prepared using a point-of-care device and was seeded onto the bottom side of DS1, DS2, and empty wounds at 80 000 cells/cm2 . Wound measurements and photographs were taken on days 0, 9, 14, 21, 28, 35, and 42 post-wounding. Histological analysis was performed on samples obtained on days 9, 14, 28, and 42. Wounds in the empty group or with ASCS alone showed increased wound contraction, fibrosis, and myofibroblast density compared with other treatment groups. The addition of ASCS to DS1 or DS2 resulted in a marked increase in re-epithelialisation of wounds at 14 days, from 15 ± 11% to 71 ± 20% (DS1 vs DS1 + ASCS) or 28 ± 14% to 77 ± 26 (DS2 vs DS2 + ASCS) despite different mechanisms of tissue regeneration employed by the DS used. These results suggest that this approach may be a viable one-stage treatment in clinical practice.
Collapse
Affiliation(s)
- Sita M Damaraju
- Product Development, Integra LifeSciences, Corp, Princeton, New Jersey, USA
| | - Benjamin R Mintz
- Product Development, Integra LifeSciences, Corp, Princeton, New Jersey, USA
| | - J Genevieve Park
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ankur Gandhi
- Product Development, Integra LifeSciences, Corp, Princeton, New Jersey, USA
| | - Sunil Saini
- Product Development, Integra LifeSciences, Corp, Princeton, New Jersey, USA
| | - Joseph A Molnar
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
74
|
Smith LA, Hidalgo Aguilar A, Owens DDG, Quelch RH, Knight E, Przyborski SA. Using Advanced Cell Culture Techniques to Differentiate Pluripotent Stem Cells and Recreate Tissue Structures Representative of Teratoma Xenografts. Front Cell Dev Biol 2021; 9:667246. [PMID: 34026759 PMCID: PMC8134696 DOI: 10.3389/fcell.2021.667246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/12/2021] [Indexed: 11/24/2022] Open
Abstract
Various methods are currently used to investigate human tissue differentiation, including human embryo culture and studies utilising pluripotent stem cells (PSCs) such as in vitro embryoid body formation and in vivo teratoma assays. Each method has its own distinct advantages, yet many are limited due to being unable to achieve the complexity and maturity of tissue structures observed in the developed human. The teratoma xenograft assay allows maturation of more complex tissue derivatives, but this method has ethical issues surrounding animal usage and significant protocol variation. In this study, we have combined three-dimensional (3D) in vitro cell technologies including the common technique of embryoid body (EB) formation with a novel porous scaffold membrane, in order to prolong cell viability and extend the differentiation of PSC derived EBs. This approach enables the formation of more complex morphologically identifiable 3D tissue structures representative of all three primary germ layers. Preliminary in vitro work with the human embryonal carcinoma line TERA2.SP12 demonstrated improved EB viability and enhanced tissue structure formation, comparable to teratocarcinoma xenografts derived in vivo from the same cell line. This is thought to be due to reduced diffusion distances as the shape of the spherical EB transforms and flattens, allowing for improved nutritional/oxygen support to the developing structures over extended periods. Further work with EBs derived from murine embryonic stem cells demonstrated that the formation of a wide range of complex, recognisable tissue structures could be achieved within 2–3 weeks of culture. Rudimentary tissue structures from all three germ layers were present, including epidermal, cartilage and epithelial tissues, again, strongly resembling tissue structure of teratoma xenografts of the same cell line. Proof of concept work with EBs derived from the human embryonic stem cell line H9 also showed the ability to form complex tissue structures within this system. This novel yet simple model offers a controllable, reproducible method to achieve complex tissue formation in vitro. It has the potential to be used to study human developmental processes, as well as offering an animal free alternative method to the teratoma assay to assess the developmental potential of novel stem cell lines.
Collapse
Affiliation(s)
- L A Smith
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - A Hidalgo Aguilar
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - D D G Owens
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - R H Quelch
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - E Knight
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - S A Przyborski
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe, NETPark, Sedgefield, United Kingdom
| |
Collapse
|
75
|
Fernández-Garibay X, Ortega MA, Cerro-Herreros E, Comelles J, Martínez E, Artero R, Fernández-Costa JM, Ramón-Azcón J. Bioengineered in vitro3D model of myotonic dystrophy type 1 human skeletal muscle. Biofabrication 2021; 13. [PMID: 33836519 DOI: 10.1088/1758-5090/abf6ae] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common hereditary myopathy in the adult population. The disease is characterized by progressive skeletal muscle degeneration that produces severe disability. At present, there is still no effective treatment for DM1 patients, but the breakthroughs in understanding the molecular pathogenic mechanisms in DM1 have allowed the testing of new therapeutic strategies. Animal models andin vitrotwo-dimensional cell cultures have been essential for these advances. However, serious concerns exist regarding how faithfully these models reproduce the biological complexity of the disease. Biofabrication tools can be applied to engineer human three-dimensional (3D) culture systems that complement current preclinical research models. Here, we describe the development of the firstin vitro3D model of DM1 human skeletal muscle. Transdifferentiated myoblasts from patient-derived fibroblasts were encapsulated in micromolded gelatin methacryloyl-carboxymethyl cellulose methacrylate hydrogels through photomold patterning on functionalized glass coverslips. These hydrogels present a microstructured topography that promotes myoblasts alignment and differentiation resulting in highly aligned myotubes from both healthy and DM1 cells in a long-lasting cell culture. The DM1 3D microtissues recapitulate the molecular alterations detected in patient biopsies. Importantly, fusion index analyses demonstrate that 3D micropatterning significantly improved DM1 cell differentiation into multinucleated myotubes compared to standard cell cultures. Moreover, the characterization of the 3D cultures of DM1 myotubes detects phenotypes as the reduced thickness of myotubes that can be used for drug testing. Finally, we evaluated the therapeutic effect of antagomiR-23b administration on bioengineered DM1 skeletal muscle microtissues. AntagomiR-23b treatment rescues both molecular DM1 hallmarks and structural phenotype, restoring myotube diameter to healthy control sizes. Overall, these new microtissues represent an improvement over conventional cell culture models and can be used as biomimetic platforms to establish preclinical studies for myotonic dystrophy.
Collapse
Affiliation(s)
- Xiomara Fernández-Garibay
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - María A Ortega
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Estefanía Cerro-Herreros
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Jordi Comelles
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain
| | - Elena Martínez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, E28029 Madrid, Spain
| | - Rubén Artero
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Juan M Fernández-Costa
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Institució Catalana de Reserca I Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, E08010 Barcelona, Spain
| |
Collapse
|
76
|
Wang S, Suhaimi H, Mabrouk M, Georgiadou S, Ward JP, Das DB. Effects of Scaffold Pore Morphologies on Glucose Transport Limitations in Hollow Fibre Membrane Bioreactor for Bone Tissue Engineering: Experiments and Numerical Modelling. MEMBRANES 2021; 11:membranes11040257. [PMID: 33918241 PMCID: PMC8065773 DOI: 10.3390/membranes11040257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
In the current research, three electrospun polycaprolactone (PCL) scaffolds with different pore morphology induced by changing the electrospinning parameters, spinning time and rate, have been prepared in order to provide a fundamental understanding on the effects pore morphology have on nutrient transport behaviour in hollow fibre membrane bioreactor (HFMB). After determining the porosity of the scaffolds, they were investigated for glucose diffusivity using cell culture media (CCM) and distilled water in a diffusion cell at 37 °C. The scanning electron microscope (SEM) images of the microstructure of the scaffolds were analysed further using ImageJ software to determine the porosity and glucose diffusivity. A Krogh cylinder model was used to determine the glucose transport profile with dimensionless variables within the HFMB. The paper discusses the roles of various dimensionless numbers (e.g., Péclet and Damköhler numbers) and non-dimensional groups of variables (e.g., non-dimensional fibre radius) on determining glucose concentration profiles, especially in the scaffold region. A negative linear relationship between glucose diffusivities across PCL scaffolds and the minimum glucose concentrations (i.e., concentration on the outer fibre edge on the outlet side (at z = 1 and r = 3.2) was also found. It was shown that the efficiency of glucose consumption improves with scaffolds of higher diffusivities. The results of this study are expected to help in optimizing designs of HFMB as well as carry out more accurate up scaling analyses for the bioreactor.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK; (S.W.); (H.S.); (M.M.); (S.G.)
| | - Hazwani Suhaimi
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK; (S.W.); (H.S.); (M.M.); (S.G.)
| | - Mostafa Mabrouk
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK; (S.W.); (H.S.); (M.M.); (S.G.)
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33El Bohouth St. (former EL Tahrir St.), Dokki, Giza P.O. Box 12622, Egypt
| | - Stella Georgiadou
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK; (S.W.); (H.S.); (M.M.); (S.G.)
| | - John P. Ward
- Department of Mathematical Sciences, Loughborough University, Loughborough LE113TU, UK;
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE113TU, UK; (S.W.); (H.S.); (M.M.); (S.G.)
- Correspondence: ; Tel.: +44-1-509-222-509
| |
Collapse
|
77
|
Shokouhimehr M, Theus AS, Kamalakar A, Ning L, Cao C, Tomov ML, Kaiser JM, Goudy S, Willett NJ, Jang HW, LaRock CN, Hanna P, Lechtig A, Yousef M, Martins JDS, Nazarian A, Harris MB, Mahmoudi M, Serpooshan V. 3D Bioprinted Bacteriostatic Hyperelastic Bone Scaffold for Damage-Specific Bone Regeneration. Polymers (Basel) 2021; 13:polym13071099. [PMID: 33808295 PMCID: PMC8036866 DOI: 10.3390/polym13071099] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Current strategies for regeneration of large bone fractures yield limited clinical success mainly due to poor integration and healing. Multidisciplinary approaches in design and development of functional tissue engineered scaffolds are required to overcome these translational challenges. Here, a new generation of hyperelastic bone (HB) implants, loaded with superparamagnetic iron oxide nanoparticles (SPIONs), are 3D bioprinted and their regenerative effect on large non-healing bone fractures is studied. Scaffolds are bioprinted with the geometry that closely correspond to that of the bone defect, using an osteoconductive, highly elastic, surgically friendly bioink mainly composed of hydroxyapatite. Incorporation of SPIONs into HB bioink results in enhanced bacteriostatic properties of bone grafts while exhibiting no cytotoxicity. In vitro culture of mouse embryonic cells and human osteoblast-like cells remain viable and functional up to 14 days on printed HB scaffolds. Implantation of damage-specific bioprinted constructs into a rat model of femoral bone defect demonstrates significant regenerative effect over the 2-week time course. While no infection, immune rejection, or fibrotic encapsulation is observed, HB grafts show rapid integration with host tissue, ossification, and growth of new bone. These results suggest a great translational potential for 3D bioprinted HB scaffolds, laden with functional nanoparticles, for hard tissue engineering applications.
Collapse
Affiliation(s)
- Mohammadreza Shokouhimehr
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Korea; (M.S.); (H.W.J.)
| | - Andrea S. Theus
- Department of Biomedical Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (M.L.T.); (N.J.W.)
| | - Archana Kamalakar
- Department of Otolaryngology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.K.); (S.G.)
| | - Liqun Ning
- Department of Biomedical Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (M.L.T.); (N.J.W.)
| | - Cong Cao
- Department of Physics, Emory University, Atlanta, GA 30322, USA;
| | - Martin L. Tomov
- Department of Biomedical Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (M.L.T.); (N.J.W.)
| | - Jarred M. Kaiser
- Department of Orthopedics, Emory University, Atlanta, GA 30322, USA;
- Atlanta Veteran’s Affairs Medical Center, Decatur, GA 30033, USA
| | - Steven Goudy
- Department of Otolaryngology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.K.); (S.G.)
| | - Nick J. Willett
- Department of Biomedical Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (M.L.T.); (N.J.W.)
- Department of Orthopedics, Emory University, Atlanta, GA 30322, USA;
- Atlanta Veteran’s Affairs Medical Center, Decatur, GA 30033, USA
| | - Ho Won Jang
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Korea; (M.S.); (H.W.J.)
| | - Christopher N. LaRock
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Philip Hanna
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (P.H.); (A.L.); (A.N.)
| | - Aron Lechtig
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (P.H.); (A.L.); (A.N.)
| | - Mohamed Yousef
- Department of Orthopedic Surgery, Sohag University, Sohag 82524, Egypt;
| | - Janaina Da Silva Martins
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, 50 Blossom St, Thier 11, Boston, MA 02114, USA;
| | - Ara Nazarian
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (P.H.); (A.L.); (A.N.)
- Department of Orthopaedic Surgery, Yerevan State Medical University, Yerevan 0025, Armenia
| | - Mitchel B. Harris
- Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Morteza Mahmoudi
- Precision Health Program & Department of Radiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Georgia Institute of Technology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (A.S.T.); (L.N.); (M.L.T.); (N.J.W.)
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
78
|
Estermann M, Spiaggia G, Septiadi D, Dijkhoff IM, Drasler B, Petri-Fink A, Rothen-Rutishauser B. Design of Perfused PTFE Vessel-Like Constructs for In Vitro Applications. Macromol Biosci 2021; 21:e2100016. [PMID: 33624920 DOI: 10.1002/mabi.202100016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Indexed: 12/18/2022]
Abstract
Tissue models mimic the complex 3D structure of human tissues, which allows the study of pathologies and the development of new therapeutic strategies. The introduction of perfusion overcomes the diffusion limitation and enables the formation of larger tissue constructs. Furthermore, it provides the possibility to investigate the effects of hematogenously administered medications. In this study, the applicability of hydrophilic polytetrafluoroethylene (PTFE) membranes as vessel-like constructs for further use in perfused tissue models is evaluated. The presented approach allows the formation of stable and leakproof tubes with a mean diameter of 654.7 µm and a wall thickness of 84.2 µm. A polydimethylsiloxane (PDMS) chip acts as a perfusion bioreactor and provides sterile conditions. As proof of concept, endothelial cells adhere to the tube's wall, express vascular endothelial cadherin (VE-cadherin) between neighboring cells, and resist perfusion at a shear rate of 0.036 N m-2 for 48 h. Furthermore, the endothelial cell layer delays significantly the diffusion of fluorescently labeled molecules into the surrounding collagen matrix and leads to a twofold reduced diffusion velocity. This approach represents a cost-effective alternative to introduce stable vessel-like constructs into tissue models, which allows adapting the surrounding matrix to the tissue properties in vivo.
Collapse
Affiliation(s)
- Manuela Estermann
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Giovanni Spiaggia
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Dedy Septiadi
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Irini Magdelina Dijkhoff
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, 1700, Switzerland.,Department of Chemistry, University of Fribourg, Chemin du Museé 9, Fribourg, 1700, Switzerland
| | | |
Collapse
|
79
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
80
|
Otto IA, Capendale PE, Garcia JP, de Ruijter M, van Doremalen RFM, Castilho M, Lawson T, Grinstaff MW, Breugem CC, Kon M, Levato R, Malda J. Biofabrication of a shape-stable auricular structure for the reconstruction of ear deformities. Mater Today Bio 2021; 9:100094. [PMID: 33665603 PMCID: PMC7903133 DOI: 10.1016/j.mtbio.2021.100094] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 11/04/2022] Open
Abstract
Bioengineering of the human auricle remains a significant challenge, where the complex and unique shape, the generation of high-quality neocartilage, and shape preservation are key factors. Future regenerative medicine–based approaches for auricular cartilage reconstruction will benefit from a smart combination of various strategies. Our approach to fabrication of an ear-shaped construct uses hybrid bioprinting techniques, a recently identified progenitor cell population, previously validated biomaterials, and a smart scaffold design. Specifically, we generated a 3D-printed polycaprolactone (PCL) scaffold via fused deposition modeling, photocrosslinked a human auricular cartilage progenitor cell–laden gelatin methacryloyl (gelMA) hydrogel within the scaffold, and cultured the bioengineered structure in vitro in chondrogenic media for 30 days. Our results show that the fabrication process maintains the viability and chondrogenic phenotype of the cells, that the compressive properties of the combined PCL and gelMA hybrid auricular constructs are similar to native auricular cartilage, and that biofabricated hybrid auricular structures exhibit excellent shape fidelity compared with the 3D digital model along with deposition of cartilage-like matrix in both peripheral and central areas of the auricular structure. Our strategy affords an anatomically enhanced auricular structure with appropriate mechanical properties, ensures adequate preservation of the auricular shape during a dynamic in vitro culture period, and enables chondrogenically potent progenitor cells to produce abundant cartilage-like matrix throughout the auricular construct. The combination of smart scaffold design with 3D bioprinting and cartilage progenitor cells holds promise for the development of clinically translatable regenerative medicine strategies for auricular reconstruction. First application of human auricular cartilage progenitor cells for bioprinting. Dual-printing of hybrid ear-shaped constructs with excellent shape fidelity over time. Strategy and design ensured adequate deposition of cartilage-like matrix throughout large auricular constructs.
Collapse
Affiliation(s)
- I A Otto
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - P E Capendale
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - J P Garcia
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - M de Ruijter
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - R F M van Doremalen
- Robotics and Mechatronics, Faculty of Electrical Engineering, Mathematics & Computer Science, University of Twente, Enschede, the Netherlands.,Bureau Science & Innovation, Deventer Hospital, Deventer, the Netherlands
| | - M Castilho
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - T Lawson
- Departments of Chemistry and Biomedical Engineering, Boston University, Boston, USA
| | - M W Grinstaff
- Departments of Chemistry and Biomedical Engineering, Boston University, Boston, USA
| | - C C Breugem
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, the Netherlands
| | - M Kon
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - R Levato
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - J Malda
- Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, the Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Science, Utrecht University, the Netherlands
| |
Collapse
|
81
|
Advances in 3D neuronal microphysiological systems: towards a functional nervous system on a chip. In Vitro Cell Dev Biol Anim 2021; 57:191-206. [PMID: 33438114 PMCID: PMC7802613 DOI: 10.1007/s11626-020-00532-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Microphysiological systems (MPS) designed to study the complexities of the peripheral and central nervous systems have made marked improvements over the years and have allowed researchers to assess in two and three dimensions the functional interconnectivity of neuronal tissues. The recent generation of brain organoids has further propelled the field into the nascent recapitulation of structural, functional, and effective connectivities which are found within the native human nervous system. Herein, we will review advances in culture methodologies, focused especially on those of human tissues, which seek to bridge the gap from 2D cultures to hierarchical and defined 3D MPS with the end goal of developing a robust nervous system-on-a-chip platform. These advances have far-reaching implications within basic science, pharmaceutical development, and translational medicine disciplines.
Collapse
|
82
|
Maharjan S, Alva J, Cámara C, Rubio AG, Hernández D, Delavaux C, Correa E, Romo MD, Bonilla D, Santiago ML, Li W, Cheng F, Ying G, Zhang YS. Symbiotic Photosynthetic Oxygenation within 3D-Bioprinted Vascularized Tissues. MATTER 2021; 4:217-240. [PMID: 33718864 PMCID: PMC7945990 DOI: 10.1016/j.matt.2020.10.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In this study, we present the photosynthetic oxygen (O2) supply to mammalian cells within a volumetric extracellular matrix-like construct, whereby a three-dimensional (3D)-bioprinted fugitive pattern encapsulating unicellular green algae, Chlamydomonas reinhardtii (C. reinhardtii), served as a natural photosynthetic O2-generator. The presence of bioprinted C. reinhardtii enhanced the viability and functionality of mammalian cells while reducing the hypoxic conditions within the tissues. We were able to subsequently endothelialize the hollow perfusable microchannels formed after enzymatic removal of the bioprinted C. reinhardtii-laden patterns from the matrices following the initial oxygenation period, to obtain biologically relevant vascularized mammalian tissue constructs. The feasibility of co-culture of C. reinhardtii with human cells, the printability and the enzymatic degradability of the fugitive bioink, as well as the exploration of C. reinhardtii as a natural, eco-friendly, cost-effective, and sustainable source of O2 would likely promote the development of engineered tissues, tissue models, and food for various applications.
Collapse
Affiliation(s)
- Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Jacqueline Alva
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Cassandra Cámara
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Andrés G. Rubio
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - David Hernández
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Clément Delavaux
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Erandy Correa
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Mariana D. Romo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Diana Bonilla
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Mille Luis Santiago
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Wanlu Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Feng Cheng
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Guoliang Ying
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
83
|
Dotson RJ, McClenahan E, Pias SC. Updated Evaluation of Cholesterol's Influence on Membrane Oxygen Permeability. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1269:23-30. [PMID: 33966190 PMCID: PMC10317081 DOI: 10.1007/978-3-030-48238-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
There is a surprising gap in knowledge regarding the mechanism of oxygen (O2) diffusional delivery at the level of tissues and cells. Yet, the effectiveness of tumor radiotherapy, the success of tissue engineering, and healthy metabolism all require ample intracellular oxygen. Tissue-level diffusion takes place in a complex and crowded macromolecular environment. Cholesterol-rich cellular membranes have been thought to reduce oxygen flux. Here, we use atomistic molecular dynamics simulations to update prior estimates of bilayer permeability and related parameters for 1-palmitoyl,2-oleoylphosphatidylcholine (POPC) and POPC/cholesterol bilayers, using a modified O2 model with improved membrane-water partitioning behavior. This work estimates an oxygen permeability coefficient of 15 ± 1 cm/s for POPC and 11.5 ± 0.4 cm/s for POPC/cholesterol (1:1 molecular ratio) at 37 °C. The permeability of POPC is found to be ~1/3 that of a water layer of similar thickness, and the permeability of POPC/cholesterol is estimated to be 20-30% below that of POPC. Void pathway visualization and free energy data support channeling of oxygen toward the center of cholesterol-incorporating membranes, while partition coefficient data suggest reduced membrane solubility of oxygen due to cholesterol. Further study is needed to understand whether diffusion pathway changes due to cholesterol and other molecular compositional factors influence oxygen availability within tissue.
Collapse
Affiliation(s)
- Rachel J Dotson
- Department of Chemistry, New Mexico Institute of Mining and Technology (New Mexico Tech), Socorro, NM, USA
| | - Emily McClenahan
- Department of Chemistry, New Mexico Institute of Mining and Technology (New Mexico Tech), Socorro, NM, USA
| | - Sally C Pias
- Department of Chemistry, New Mexico Institute of Mining and Technology (New Mexico Tech), Socorro, NM, USA.
| |
Collapse
|
84
|
Thorp H, Kim K, Kondo M, Grainger DW, Okano T. Fabrication of hyaline-like cartilage constructs using mesenchymal stem cell sheets. Sci Rep 2020; 10:20869. [PMID: 33257787 PMCID: PMC7705723 DOI: 10.1038/s41598-020-77842-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/10/2020] [Indexed: 12/21/2022] Open
Abstract
Cell and tissue engineering approaches for articular cartilage regeneration increasingly focus on mesenchymal stem cells (MSCs) as allogeneic cell sources, based on availability and innate chondrogenic potential. Many MSCs exhibit chondrogenic potential as three-dimensional (3D) cultures (i.e. pellets and seeded biomaterial scaffolds) in vitro; however, these constructs present engraftment, biocompatibility, and cell functionality limitations in vivo. Cell sheet technology maintains cell functionality as scaffold-free constructs while enabling direct cell transplantation from in vitro culture to targeted sites in vivo. The present study aims to develop transplantable hyaline-like cartilage constructs by stimulating MSC chondrogenic differentiation as cell sheets. To achieve this goal, 3D MSC sheets are prepared, exploiting spontaneous post-detachment cell sheet contraction, and chondrogenically induced. Results support 3D MSC sheets' chondrogenic differentiation to hyaline cartilage in vitro via post-contraction cytoskeletal reorganization and structural transformations. These 3D cell sheets' initial thickness and cellular densities may also modulate MSC-derived chondrocyte hypertrophy in vitro. Furthermore, chondrogenically differentiated cell sheets adhere directly to cartilage surfaces via retention of adhesion molecules while maintaining the cell sheets' characteristics. Together, these data support the utility of cell sheet technology for fabricating scaffold-free, hyaline-like cartilage constructs from MSCs for future transplantable articular cartilage regeneration therapies.
Collapse
Affiliation(s)
- Hallie Thorp
- Department of Pharmaceutics and Pharmaceutical Chemistry, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, 30 South 2000 East, Salt Lake City, UT, 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Kyungsook Kim
- Department of Pharmaceutics and Pharmaceutical Chemistry, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, 30 South 2000 East, Salt Lake City, UT, 84112, USA.
| | - Makoto Kondo
- Department of Pharmaceutics and Pharmaceutical Chemistry, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, 30 South 2000 East, Salt Lake City, UT, 84112, USA
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, 30 South 2000 East, Salt Lake City, UT, 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Teruo Okano
- Department of Pharmaceutics and Pharmaceutical Chemistry, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, 30 South 2000 East, Salt Lake City, UT, 84112, USA.
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
85
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
86
|
Paez-Mayorga J, Capuani S, Hernandez N, Farina M, Chua CYX, Blanchard R, Sizovs A, Liu HC, Fraga DW, Niles JA, Salazar HF, Corradetti B, Sikora AG, Kloc M, Li XC, Gaber AO, Nichols JE, Grattoni A. Neovascularized implantable cell homing encapsulation platform with tunable local immunosuppressant delivery for allogeneic cell transplantation. Biomaterials 2020; 257:120232. [DOI: 10.1016/j.biomaterials.2020.120232] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 01/10/2023]
|
87
|
Robering JW, Al-Abboodi M, Titzmann A, Horn I, Beier JP, Horch RE, Kengelbach-Weigand A, Boos AM. Tissue Engineering of Lymphatic Vasculature in the Arteriovenous Loop Model of the Rat. Tissue Eng Part A 2020; 27:129-141. [PMID: 32524901 DOI: 10.1089/ten.tea.2020.0108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Various therapeutic approaches, for example, in case of trauma or cancer require the transplantation of autologous tissue. Depending on the size and the origin of the harvested tissue, these therapies can lead to iatrogenic complications and donor-site morbidities. In future, these side effects could be avoided by transplanting artificially generated tissue consisting of different cell types and matrix components derived from the host body. Tissue that is grown in the patient could be advantageous compared with the more simply structured in vitro-grown alternatives. To overcome the limitations of graft vascularization, the arteriovenous (AV) loop technique has been established for different tissues in the last years and was adapted for lymphatic tissue engineering in the present study. We utilized the AV loop technique to grow human lymphatic vasculature in vivo in the Rowett nude (RNU) rat. A combination of human lymphatic endothelial cells (LECs) and bone marrow-derived mesenchymal stem cells was implanted in a fibrin matrix surrounding the AV loop. After 2 or 4 weeks of implantation, the animals were perfused and the tissue was harvested. It could be demonstrated by immunohistochemistry for human LYVE1, human CD31, and murine podoplanin that the implanted cells formed human lymphatic vasculature in the AV loop chamber. Beside development of murine podoplanin-positive vasculature in the AV loop tissue, vasculature positive for human marker proteins developed in comparable numbers. This suggests that implanted LECs are able to improve the lymphatic vascularization of the newly engineered tissue. Thus, we were able to establish an in vivo tissue engineering method to generate lymphatic vascularized soft tissue. An axially vascularized transplantable lymphatic vessel network was engineered without requiring advanced cell culture equipment, rendering the lymphatic AV loop highly suitable for applied regenerative medicine. Impact statement Various surgical procedures require the transplantation of autologous harvested tissue, for example, the vascularized lymph node transfer for the treatment of lymphedema. Tissue-engineered transplants could be used instead of autologous transplants and thereby help to reduce the side effects of those therapies. However, in vitro tissue engineering of large constructs requires a lot of know-how as well as advanced cell culture equipment, which might not be accessible in every hospital. In vivo tissue engineering approaches like the presented technique for the generation of transplantable networks of lymphatic vasculature could serve as an alternative for in vitro tissue engineering approaches in clinical settings.
Collapse
Affiliation(s)
- Jan W Robering
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany.,Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Majida Al-Abboodi
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany.,Institute of Genetic Engineering and Biotechnology, University of Baghdad, Baghdad, Iraq
| | - Adriana Titzmann
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany
| | - Inge Horn
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany
| | - Justus P Beier
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany.,Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany
| | - Anja M Boos
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital, Erlangen, Germany.,Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
88
|
Patino-Guerrero A, Veldhuizen J, Zhu W, Migrino RQ, Nikkhah M. Three-dimensional scaffold-free microtissues engineered for cardiac repair. J Mater Chem B 2020; 8:7571-7590. [PMID: 32724973 PMCID: PMC8314954 DOI: 10.1039/d0tb01528h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases, including myocardial infarction (MI), persist as the leading cause of mortality and morbidity worldwide. The limited regenerative capacity of the myocardium presents significant challenges specifically for the treatment of MI and, subsequently, heart failure (HF). Traditional therapeutic approaches mainly rely on limiting the induced damage or the stress on the remaining viable myocardium through pharmacological regulation of remodeling mechanisms, rather than replacement or regeneration of the injured tissue. The emerging alternative regenerative medicine-based approaches have focused on restoring the damaged myocardial tissue with newly engineered functional and bioinspired tissue units. Cardiac regenerative medicine approaches can be broadly categorized into three groups: cell-based therapies, scaffold-based cardiac tissue engineering, and scaffold-free cardiac tissue engineering. Despite significant advancements, however, the clinical translation of these approaches has been critically hindered by two key obstacles for successful structural and functional replacement of the damaged myocardium, namely: poor engraftment of engineered tissue into the damaged cardiac muscle and weak electromechanical coupling of transplanted cells with the native tissue. To that end, the integration of micro- and nanoscale technologies along with recent advancements in stem cell technologies have opened new avenues for engineering of structurally mature and highly functional scaffold-based (SB-CMTs) and scaffold-free cardiac microtissues (SF-CMTs) with enhanced cellular organization and electromechanical coupling for the treatment of MI and HF. In this review article, we will present the state-of-the-art approaches and recent advancements in the engineering of SF-CMTs for myocardial repair.
Collapse
|
89
|
Ichanti H, Sladic S, Kalies S, Haverich A, Andrée B, Hilfiker A. Characterization of Tissue Engineered Endothelial Cell Networks in Composite Collagen-Agarose Hydrogels. Gels 2020; 6:gels6030027. [PMID: 32899293 PMCID: PMC7559300 DOI: 10.3390/gels6030027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
Scaffolds constitute an important element in vascularized tissues and are therefore investigated for providing the desired mechanical stability and enabling vasculogenesis and angiogenesis. In this study, supplementation of hydrogels containing either MatrigelTM and rat tail collagen I (MatrigelTM/rCOL) or human collagen (hCOL) with SeaPlaqueTM agarose were analyzed with regard to construct thickness and formation and characteristics of endothelial cell (EC) networks compared to constructs without agarose. Additionally, the effect of increased rCOL content in MatrigelTM/rCOL constructs was studied. An increase of rCOL content from 1 mg/mL to 3 mg/mL resulted in an increase of construct thickness by approximately 160%. The high rCOL content, however, impaired the formation of an EC network. The supplementation of MatrigelTM/rCOL with agarose increased the thickness of the hydrogel construct by approximately 100% while supporting the formation of a stable EC network. The use of hCOL/agarose composite hydrogels led to a slight increase in the thickness of the 3D hydrogel construct and supported the formation of a multi-layered EC network compared to control constructs. Our findings suggest that agarose/collagen-based composite hydrogels are promising candidates for tissue engineering of vascularized constructs as cell viability is maintained and the formation of a stable and multi-layered EC network is supported.
Collapse
Affiliation(s)
- Houda Ichanti
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
| | - Sanja Sladic
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
| | - Stefan Kalies
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany;
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, 30625 Hannover, Germany
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
| | - Birgit Andrée
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
- Correspondence: (B.A.); (A.H.); Tel.: +49-511-532-8913 (B.A.); +49-511-532-8998 (A.H.)
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
- Correspondence: (B.A.); (A.H.); Tel.: +49-511-532-8913 (B.A.); +49-511-532-8998 (A.H.)
| |
Collapse
|
90
|
Xiao W, Qu X, Tan Y, Xiao J, Le Y, Li Y, Liu X, Li B, Liao X. Synthesis of photocrosslinkable hydrogels for engineering three-dimensional vascular-like constructs by surface tension-driven assembly. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111143. [PMID: 32806229 DOI: 10.1016/j.msec.2020.111143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/13/2020] [Accepted: 05/28/2020] [Indexed: 01/01/2023]
Abstract
Surface tension-driven assembly is a simple routine used in modular tissue engineering to create three-dimensional (3D) biomimetic tissues with desired structural and biological characteristics. A major bottleneck for this technology is the lack of suitable hydrogel materials to meet the requirements of the assembly process and tissue regeneration. Identifying specific requirements and synthesizing novel hydrogels will provide a versatile platform for generating additional biomimetic functional tissues using this approach. In this paper, we present a novel composite hydrogel system based on methacrylated gelatin and γ-polyglutamic acid by UV copolymerization as the building block for fabricating vascular-like tissue via surface tension-driven assembly. The resulting composite hydrogels exhibited the improved mechanical properties and hydrophilicity, which greatly facilitate the assembly process. Subsequent cell encapsulation experiment proved that the hydrogel could provide 3D support for cellular spreading and migration. Furthermore, based on the composite microgel building blocks, cylindrical vascular-like construct with a perfusable microchannel was generated by the needle-assisted sequential assembly. In order to construct a biomimetic vascular tissue, the endothelial cells and smooth muscle cells were encapsulated in the microgels assembly with a spatial arrangement to build a heterogeneous double-layer tubular structure and the cells could readily elongate and migrate in the hollow concentric construct over 3 days. These data suggest that this composite hydrogel is an attractive candidate for surface tension-driven assembly purposes, making the hydrogel potentially applicable in the fabrication of biomimetic vascularized tissues.
Collapse
Affiliation(s)
- Wenqian Xiao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Xiaohang Qu
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Yunfei Tan
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Jing Xiao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Yinpeng Le
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Yongxiang Li
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Xue Liu
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Bo Li
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China.
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China.
| |
Collapse
|
91
|
Cartaxo AL, Estrada MF, Domenici G, Roque R, Silva F, Gualda EJ, Loza-Alvarez P, Sflomos G, Brisken C, Alves PM, André S, Brito C. A novel culture method that sustains ERα signaling in human breast cancer tissue microstructures. J Exp Clin Cancer Res 2020; 39:161. [PMID: 32807212 PMCID: PMC7430012 DOI: 10.1186/s13046-020-01653-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Estrogen receptor α (ERα) signaling is a defining and driving event in most breast cancers; ERα is detected in malignant epithelial cells of 75% of all breast cancers (classified as ER-positive breast cancer) and, in these cases, ERα targeting is the main therapeutic strategy. However, the biological determinants of ERα heterogeneity and the mechanisms underlying therapeutic resistance are still elusive, hampered by the challenges in developing experimental models recapitulative of intra-tumoral heterogeneity and in which ERα signaling is sustained. Ex vivo cultures of human breast cancer tissue have been proposed to retain the original tissue architecture, epithelial and stromal cell components and ERα. However, loss of cellularity, viability and ERα expression are well-known culture-related phenomena. METHODS BC samples were collected and brought to the laboratory. Then they were minced, enzymatically digested, entrapped in alginate and cultured for 1 month. The histological architecture, cellular composition and cell proliferation of tissue microstructures were assessed by immunohistochemistry. Cell viability was assessed by measurement of cell metabolic activity and histological evaluation. The presence of ERα was accessed by immunohistochemistry and RT-qPCR and its functionality evaluated by challenge with 17-β-estradiol and fulvestrant. RESULTS We describe a strategy based on entrapment of breast cancer tissue microstructures in alginate capsules and their long-term culture under agitation, successfully applied to tissue obtained from 63 breast cancer patients. After 1 month in culture, the architectural features of the encapsulated tissue microstructures were similar to the original patient tumors: epithelial, stromal and endothelial compartments were maintained, with an average of 97% of cell viability compared to day 0. In ERα-positive cases, fibers of collagen, the main extracellular matrix component in vivo, were preserved. ERα expression was at least partially retained at gene and protein levels and response to ERα stimulation and inhibition was observed at the level of downstream targets, demonstrating active ER signaling. CONCLUSIONS The proposed model system is a new methodology to study ex vivo breast cancer biology, in particular ERα signaling. It is suitable for interrogating the long-term effects of anti-endocrine drugs in a set-up that closely resembles the original tumor microenvironment, with potential application in pre- and co-clinical assays of ERα-positive breast cancer.
Collapse
Affiliation(s)
- Ana Luísa Cartaxo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Oeiras, Portugal
| | - Marta F Estrada
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Oeiras, Portugal
| | - Giacomo Domenici
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Oeiras, Portugal
| | - Ruben Roque
- IPOLFG, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Fernanda Silva
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Emilio J Gualda
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Pablo Loza-Alvarez
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - George Sflomos
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Oeiras, Portugal
| | - Saudade André
- IPOLFG, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Oeiras, Portugal.
| |
Collapse
|
92
|
Dikici S, Claeyssens F, MacNeil S. Pre-Seeding of Simple Electrospun Scaffolds with a Combination of Endothelial Cells and Fibroblasts Strongly Promotes Angiogenesis. Tissue Eng Regen Med 2020; 17:445-458. [PMID: 32447555 PMCID: PMC7392995 DOI: 10.1007/s13770-020-00263-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/14/2020] [Accepted: 04/08/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Introduction of pro-angiogenic cells into tissue-engineered (TE) constructs (prevascularisation) is a promising approach to overcome delayed neovascularisation of such constructs post-implantation. Accordingly, in this study, we examined the contribution of human dermal microvascular endothelial cells (HDMECs) and human dermal fibroblasts (HDFs) alone and in combination on the formation of new blood vessels in ex-ovo chick chorioallantoic membrane (CAM) assay. METHODS Poly-3-hydroxybutyrate-co-3-hydroxyvalerate (PHBV) and polycaprolactone (PCL) were first examined in terms of their physical, mechanical, and biological performances. The effect of gelatin coating and co-culture conditions on enhancing endothelial cell viability and growth was then investigated. Finally, the angiogenic potential of HDMECs and HDFs were assessed macroscopically and histologically after seeding on simple electrospun PHBV scaffolds either in isolation or in indirect co-culture using an ex-ovo CAM assay. RESULTS The results demonstrated that PHBV was slightly more favourable than PCL for HDMECs in terms of cell metabolic activity. The gelatin coating of PHBV scaffolds and co-culture of HDMECs with HDFs both showed a positive impact on HDMECs viability and growth. Both cell types induced angiogenesis over 7 days in the CAM assay either in isolation or in co-culture. The introduction of HDMECs to the scaffolds resulted in the production of more blood vessels in the area of implantation than the introduction of HDFs, but the co-culture of HDMECs and HDFs gave the most significant angiogenic activity. CONCLUSION Our findings showed that the in vitro prevascularisation of TE constructs with HDMECs and HDFs alone or in co-culture promotes angiogenesis in implantable TE constructs.
Collapse
Affiliation(s)
- Serkan Dikici
- Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, North Campus Broad Lane, Sheffield, S3 7HQ, UK
| | - Frederik Claeyssens
- Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, North Campus Broad Lane, Sheffield, S3 7HQ, UK
| | - Sheila MacNeil
- Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, North Campus Broad Lane, Sheffield, S3 7HQ, UK.
| |
Collapse
|
93
|
Calar K, Plesselova S, Bhattacharya S, Jorgensen M, de la Puente P. Human Plasma-Derived 3D Cultures Model Breast Cancer Treatment Responses and Predict Clinically Effective Drug Treatment Concentrations. Cancers (Basel) 2020; 12:cancers12071722. [PMID: 32610529 PMCID: PMC7407241 DOI: 10.3390/cancers12071722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023] Open
Abstract
Lack of efficacy and a low overall success rate of phase I-II clinical trials are the most common failures when it comes to advancing cancer treatment. Current drug sensitivity screenings present several challenges including differences in cell growth rates, the inconsistent use of drug metrics, and the lack of translatability. Here, we present a patient-derived 3D culture model to overcome these limitations in breast cancer (BCa). The human plasma-derived 3D culture model (HuP3D) utilizes patient plasma as the matrix, where BCa cell lines and primary BCa biopsies were grown and screened for drug treatments. Several drug metrics were evaluated from relative cell count and growth rate curves. Correlations between HuP3D metrics, established preclinical models, and clinical effective concentrations in patients were determined. HuP3D efficiently supported the growth and expansion of BCa cell lines and primary breast cancer tumors as both organoids and single cells. Significant and strong correlations between clinical effective concentrations in patients were found for eight out of ten metrics for HuP3D, while a very poor positive correlation and a moderate correlation was found for 2D models and other 3D models, respectively. HuP3D is a feasible and efficacious platform for supporting the growth and expansion of BCa, allowing high-throughput drug screening and predicting clinically effective therapies better than current preclinical models.
Collapse
Affiliation(s)
- Kristin Calar
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
| | - Simona Plesselova
- Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain;
| | - Somshuvra Bhattacharya
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
| | - Megan Jorgensen
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
- MD/PhD Program, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA
| | - Pilar de la Puente
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA; (K.C.); (S.B.); (M.J.)
- Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, USA
- Flow Cytometry Core, Sanford Research, Sioux Falls, SD 57104, USA
- Correspondence: ; Tel.: +1-605-312-6042
| |
Collapse
|
94
|
Melke J, Zhao F, Ito K, Hofmann S. Orbital seeding of mesenchymal stromal cells increases osteogenic differentiation and bone-like tissue formation. J Orthop Res 2020; 38:1228-1237. [PMID: 31922286 PMCID: PMC7317919 DOI: 10.1002/jor.24583] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 12/12/2019] [Indexed: 02/04/2023]
Abstract
In bone tissue engineering (TE), an efficient seeding and homogenous distribution of cells is needed to avoid cell loss and damage as well as to facilitate tissue development. Dynamic seeding methods seem to be superior to the static ones because they tend to result in a more homogeneous cell distribution by using kinetic forces. However, most dynamic seeding techniques are elaborate or require special equipment and its influence on the final bone tissue-engineered construct is not clear. In this study, we applied a simple, dynamic seeding method using an orbital shaker to seed human bone marrow-derived mesenchymal stromal cells (hBMSCs) on silk fibroin scaffolds. Significantly higher cell numbers with a more homogenous cell distribution, increased osteogenic differentiation, and mineral deposition were observed using the dynamic approach both for 4 and 6 hours as compared to the static seeding method. The positive influence of dynamic seeding could be attributed to both cell density and distribution but also nutrient supply during seeding and shear stresses (0.0-3.0 mPa) as determined by computational simulations. The influence of relevant mechanical stimuli during seeding should be investigated in the future, especially regarding the importance of mechanical cues for bone TE applications. Our results highlight the importance of adequate choice of seeding method and its impact on developing tissue-engineered constructs. The application of this simple seeding technique is not only recommended for bone TE but can also be used for seeding similar porous scaffolds with hBMSCs in other TE fields.
Collapse
Affiliation(s)
- Johanna Melke
- Orthopaedic BiomechanicsDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands,Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| | - Feihu Zhao
- Orthopaedic BiomechanicsDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands,Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| | - Keita Ito
- Orthopaedic BiomechanicsDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands,Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| | - Sandra Hofmann
- Orthopaedic BiomechanicsDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands,Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| |
Collapse
|
95
|
Tasnim F, Singh NH, Tan EKF, Xing J, Li H, Hissette S, Manesh S, Fulwood J, Gupta K, Ng CW, Xu S, Hill J, Yu H. Tethered primary hepatocyte spheroids on polystyrene multi-well plates for high-throughput drug safety testing. Sci Rep 2020; 10:4768. [PMID: 32179810 PMCID: PMC7075904 DOI: 10.1038/s41598-020-61699-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 02/19/2020] [Indexed: 12/26/2022] Open
Abstract
Hepatocyte spheroids are useful models for mimicking liver phenotypes in vitro because of their three-dimensionality. However, the lack of a biomaterial platform which allows the facile manipulation of spheroid cultures on a large scale severely limits their application in automated high-throughput drug safety testing. In addition, there is not yet a robust way of controlling spheroid size, homogeneity and integrity during extended culture. This work addresses these bottlenecks to the automation of hepatocyte spheroid culture by tethering 3D hepatocyte spheroids directly onto surface-modified polystyrene (PS) multi-well plates. However, polystyrene surfaces are inert toward functionalization, and this makes the uniform conjugation of bioactive ligands very challenging. Surface modification of polystyrene well plates is achieved herein using a three-step sequence, resulting in a homogeneous distribution of bioactive RGD and galactose ligands required for spheroid tethering and formation. Importantly, treatment of polystyrene tethered spheroids with vehicle and paradigm hepatotoxicant (chlorpromazine) treatment using an automated liquid handling platform shows low signal deviation, intact 3D spheroidal morphology and Z’ values above 0.5, and hence confirming their amenability to high-throughput automation. Functional analyses performance (i.e. urea and albumin production, cytochrome P450 activity and induction studies) of the polystyrene tethered spheroids reveal significant improvements over hepatocytes cultured as collagen monolayers. This is the first demonstration of automated hepatotoxicant treatment on functional 3D hepatocyte spheroids tethered directly on polystyrene multi-well plates, and will serve as an important advancement in the application of 3D tethered spheroid models to high throughput drug screening.
Collapse
Affiliation(s)
- Farah Tasnim
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Nisha Hari Singh
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Elijah Keng Foo Tan
- Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Yong Loo Lin School of Medicine (Department of Physiology) and Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
| | - Jiangwa Xing
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Huan Li
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Sebastien Hissette
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Sravanthy Manesh
- Experimental Therapeutics Centre (ETC), Level 3, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Justina Fulwood
- Experimental Therapeutics Centre (ETC), Level 3, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Kapish Gupta
- Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Chan Way Ng
- Yong Loo Lin School of Medicine (Department of Physiology) and Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
| | - Shuoyu Xu
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Jeffrey Hill
- Experimental Therapeutics Centre (ETC), Level 3, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore.,Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton, BN19RH, UK
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore. .,Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore. .,Yong Loo Lin School of Medicine (Department of Physiology) and Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore. .,CAMP IRG, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Enterprise Wing, Level 4, Singapore, 138602, Singapore.
| |
Collapse
|
96
|
Mohamed MGA, Ambhorkar P, Samanipour R, Yang A, Ghafoor A, Kim K. Microfluidics-based fabrication of cell-laden microgels. BIOMICROFLUIDICS 2020; 14:021501. [PMID: 32161630 PMCID: PMC7058428 DOI: 10.1063/1.5134060] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/16/2020] [Indexed: 05/02/2023]
Abstract
Microfluidic principles have been extensively utilized as powerful tools to fabricate controlled monodisperse cell-laden hydrogel microdroplets for various biological applications, especially tissue engineering. In this review, we report recent advances in microfluidic-based droplet fabrication and provide our rationale to justify the superiority of microfluidics-based techniques over other microtechnology methods in achieving the encapsulation of cells within hydrogels. The three main components of such a system-hydrogels, cells, and device configurations-are examined thoroughly. First, the characteristics of various types of hydrogels including natural and synthetic types, especially concerning cell encapsulation, are examined. This is followed by the elucidation of the reasoning behind choosing specific cells for encapsulation. Next, in addition to a detailed discussion of their respective droplet formation mechanisms, various device configurations including T-junctions, flow-focusing, and co-flowing that aid in achieving cell encapsulation are critically reviewed. We then present an outlook on the current applications of cell-laden hydrogel droplets in tissue engineering such as 3D cell culturing, rapid generation and repair of tissues, and their usage as platforms for studying cell-cell and cell-microenvironment interactions. Finally, we shed some light upon the prospects of microfluidics-based production of cell-laden microgels and propose some directions for forthcoming research that can aid in overcoming challenges currently impeding the translation of the technology into clinical success.
Collapse
Affiliation(s)
- Mohamed G. A. Mohamed
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Pranav Ambhorkar
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Roya Samanipour
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Annie Yang
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Ali Ghafoor
- Irving K. Barber School of Arts and Sciences, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | | |
Collapse
|
97
|
Sieberath A, Della Bella E, Ferreira AM, Gentile P, Eglin D, Dalgarno K. A Comparison of Osteoblast and Osteoclast In Vitro Co-Culture Models and Their Translation for Preclinical Drug Testing Applications. Int J Mol Sci 2020; 21:E912. [PMID: 32019244 PMCID: PMC7037207 DOI: 10.3390/ijms21030912] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
As the population of western societies on average ages, the number of people affected by bone remodeling-associated diseases such as osteoporosis continues to increase. The development of new therapeutics is hampered by the high failure rates of drug candidates during clinical testing, which is in part due to the poor predictive character of animal models during preclinical drug testing. Co-culture models of osteoblasts and osteoclasts offer an alternative to animal testing and are considered to have the potential to improve drug development processes in the future. However, a robust, scalable, and reproducible 3D model combining osteoblasts and osteoclasts for preclinical drug testing purposes has not been developed to date. Here we review various types of osteoblast-osteoclast co-culture models and outline the remaining obstacles that must be overcome for their successful translation.
Collapse
Affiliation(s)
- Alexander Sieberath
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| | - Elena Della Bella
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland; (E.D.B.); (D.E.)
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland; (E.D.B.); (D.E.)
| | - Kenny Dalgarno
- School of Engineering, Newcastle University, Newcastle-Upon-Tyne NE1 7RU, UK; (A.S.); (A.M.F.); (P.G.)
| |
Collapse
|
98
|
Bargavi P, Ramya R, Chitra S, Vijayakumari S, Riju Chandran R, Durgalakshmi D, Rajashree P, Balakumar S. Bioactive, degradable and multi-functional three-dimensional membranous scaffolds of bioglass and alginate composites for tissue regenerative applications. Biomater Sci 2020; 8:4003-4025. [DOI: 10.1039/d0bm00714e] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multifunctional bioactive hydrogel ECM like membrane for 3D dynamic tissue/disease modelling.
Collapse
Affiliation(s)
- P. Bargavi
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600 025
- India
| | - R. Ramya
- SRM Dental College
- SRMIST
- Chennai – 600089
- India
| | - S. Chitra
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600 025
- India
| | - S. Vijayakumari
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600 025
- India
| | - R. Riju Chandran
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600 025
- India
| | - D. Durgalakshmi
- Department of Medical Physics
- Anna University
- Chennai – 600 025
- India
| | - P. Rajashree
- CAS in Crystallography & Biophysics
- University of Madras
- Chennai – 600 025
- India
| | - S. Balakumar
- National Centre for Nanoscience and Nanotechnology
- University of Madras
- Chennai – 600 025
- India
| |
Collapse
|
99
|
Müller WEG, Schepler H, Tolba E, Wang S, Ackermann M, Muñoz-Espí R, Xiao S, Tan R, She Z, Neufurth M, Schröder HC, Wang X. A physiologically active interpenetrating collagen network that supports growth and migration of epidermal keratinocytes: zinc-polyP nanoparticles integrated into compressed collagen. J Mater Chem B 2020; 8:5892-5902. [DOI: 10.1039/d0tb01240h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It is demonstrated that polyphosphate, as a component in wound healing mats together with Zn2+, is essential for growth and migration of skin keratinocytes.
Collapse
|
100
|
Saffari TM, Bedar M, Hundepool CA, Bishop AT, Shin AY. The role of vascularization in nerve regeneration of nerve graft. Neural Regen Res 2020; 15:1573-1579. [PMID: 32209756 PMCID: PMC7437604 DOI: 10.4103/1673-5374.276327] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Vascularization is an important factor in nerve graft survival and function. The specific molecular regulations and patterns of angiogenesis following peripheral nerve injury are in a broad complex of pathways. This review aims to summarize current knowledge on the role of vascularization in nerve regeneration, including the key regulation molecules, and mechanisms and patterns of revascularization after nerve injury. Angiogenesis, the maturation of pre-existing vessels into new areas, is stimulated through angiogenic factors such as vascular endothelial growth factor and precedes the repair of damaged nerves. Vascular endothelial growth factor administration to nerves has demonstrated to increase revascularization after injury in basic science research. In the clinical setting, vascularized nerve grafts could be used in the reconstruction of large segmental peripheral nerve injuries. Vascularized nerve grafts are postulated to accelerate revascularization and enhance nerve regeneration by providing an optimal nutritional environment, especially in scarred beds, and decrease fibroblast infiltration. This could improve functional recovery after nerve grafting, however, conclusive evidence of the superiority of vascularized nerve grafts is lacking in human studies. A well-designed randomized controlled trial comparing vascularized nerve grafts to non-vascularized nerve grafts involving patients with similar injuries, nerve graft repair and follow-up times is necessary to demonstrate the efficacy of vascularized nerve grafts. Due to technical challenges, composite transfer of a nerve graft along with its adipose tissue has been proposed to provide a healthy tissue bed. Basic science research has shown that a vascularized fascial flap containing adipose tissue and a vascular bundle improves revascularization through excreted angiogenic factors, provided by the stem cells in the adipose tissue as well as by the blood supply and environmental support. While it was previously believed that revascularization occurred from both nerve ends, recent studies propose that revascularization occurs primarily from the proximal nerve coaptation. Fascial flaps or vascularized nerve grafts have limited applicability and future directions could lead towards off-the-shelf alternatives to autografting, such as biodegradable nerve scaffolds which include capillary-like networks to enable vascularization and avoid graft necrosis and ischemia.
Collapse
Affiliation(s)
- Tiam M Saffari
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Meiwand Bedar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Caroline A Hundepool
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Allen T Bishop
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Alexander Y Shin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|