101
|
Chae S, Hong J, Hwangbo H, Kim G. The utility of biomedical scaffolds laden with spheroids in various tissue engineering applications. Am J Cancer Res 2021; 11:6818-6832. [PMID: 34093855 PMCID: PMC8171099 DOI: 10.7150/thno.58421] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
A spheroid is a complex, spherical cellular aggregate supporting cell-cell and cell-matrix interactions in an environment that mimics the real-world situation. In terms of tissue engineering, spheroids are important building blocks that replace two-dimensional cell cultures. Spheroids replicate tissue physiological activities. The use of spheroids with/without scaffolds yields structures that engage in desired activities and replicate the complicated geometry of three-dimensional tissues. In this mini-review, we describe conventional and novel methods by which scaffold-free and scaffolded spheroids may be fabricated and discuss their applications in tissue regeneration and future perspectives.
Collapse
|
102
|
Correia CR, Bjørge IM, Nadine S, Mano JF. Minimalist Tissue Engineering Approaches Using Low Material-Based Bioengineered Systems. Adv Healthc Mater 2021; 10:e2002110. [PMID: 33709572 DOI: 10.1002/adhm.202002110] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/17/2021] [Indexed: 12/14/2022]
Abstract
From an "over-engineering" era in which biomaterials played a central role, now it is observed to the emergence of "developmental" tissue engineering (TE) strategies which rely on an integrative cell-material perspective that paves the way for cell self-organization. The current challenge is to engineer the microenvironment without hampering the spontaneous collective arrangement ability of cells, while simultaneously providing biochemical, geometrical, and biophysical cues that positively influence tissue healing. These efforts have resulted in the development of low-material based TE strategies focused on minimizing the amount of biomaterial provided to the living key players of the regenerative process. Through a "minimalist-engineering" approach, the main idea is to fine-tune the spatial balance occupied by the inanimate region of the regenerative niche toward maximum actuation of the key living components during the healing process.
Collapse
Affiliation(s)
- Clara R. Correia
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| | - Isabel M. Bjørge
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| | - Sara Nadine
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| | - João F. Mano
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| |
Collapse
|
103
|
Decante G, Costa JB, Silva-Correia J, Collins MN, Reis RL, Oliveira JM. Engineering bioinks for 3D bioprinting. Biofabrication 2021; 13. [PMID: 33662949 DOI: 10.1088/1758-5090/abec2c] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
In recent years, three-dimensional (3D) bioprinting has attracted wide research interest in biomedical engineering and clinical applications. This technology allows for unparalleled architecture control, adaptability and repeatability that can overcome the limits of conventional biofabrication techniques. Along with the emergence of a variety of 3D bioprinting methods, bioinks have also come a long way. From their first developments to support bioprinting requirements, they are now engineered to specific injury sites requirements to mimic native tissue characteristics and to support biofunctionality. Current strategies involve the use of bioinks loaded with cells and biomolecules of interest, without altering their functions, to deliverin situthe elements required to enhance healing/regeneration. The current research and trends in bioink development for 3D bioprinting purposes is overviewed herein.
Collapse
Affiliation(s)
- Guy Decante
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João B Costa
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maurice N Collins
- Bernal Institute, School of Engineering, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
104
|
Recent advances in bioprinting technologies for engineering different cartilage-based tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112005. [PMID: 33812625 DOI: 10.1016/j.msec.2021.112005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Inadequate self-repair and regenerative efficiency of the cartilage tissues has motivated the researchers to devise advanced and effective strategies to resolve this issue. Introduction of bioprinting to tissue engineering has paved the way for fabricating complex biomimetic engineered constructs. In this context, the current review gears off with the discussion of standard and advanced 3D/4D printing technologies and their implications for the repair of different cartilage tissues, namely, articular, meniscal, nasoseptal, auricular, costal, and tracheal cartilage. The review is then directed towards highlighting the current stem cell opportunities. On a concluding note, associated critical issues and prospects for future developments, particularly in this sphere of personalized medicines have been discussed.
Collapse
|
105
|
Quadri F, Soman SS, Vijayavenkataraman S. Progress in cardiovascular bioprinting. Artif Organs 2021; 45:652-664. [PMID: 33432583 DOI: 10.1111/aor.13913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/13/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease has been the leading cause of death globally for the past 15 years. Following a major cardiac disease episode, the ideal treatment would be the replacement of the damaged tissue, due to the limited regenerative capacity of cardiac tissues. However, we suffer from a chronic organ donor shortage which causes approximately 20 people to die each day waiting to receive an organ. Bioprinting of tissues and organs can potentially alleviate this burden by fabricating low cost tissue and organ replacements for cardiac patients. Clinical adoption of bioprinting in cardiovascular medicine is currently limited by the lack of systematic demonstration of its effectiveness, high costs, and the complexity of the workflow. Here, we give a concise review of progress in cardiovascular bioprinting and its components. We further discuss the challenges and future prospects of cardiovascular bioprinting in clinical applications.
Collapse
Affiliation(s)
- Faisal Quadri
- Division of Science, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Soja Saghar Soman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Sanjairaj Vijayavenkataraman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, USA
| |
Collapse
|
106
|
Benwood C, Chrenek J, Kirsch RL, Masri NZ, Richards H, Teetzen K, Willerth SM. Natural Biomaterials and Their Use as Bioinks for Printing Tissues. Bioengineering (Basel) 2021; 8:27. [PMID: 33672626 PMCID: PMC7924193 DOI: 10.3390/bioengineering8020027] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The most prevalent form of bioprinting-extrusion bioprinting-can generate structures from a diverse range of materials and viscosities. It can create personalized tissues that aid in drug testing and cancer research when used in combination with natural bioinks. This paper reviews natural bioinks and their properties and functions in hard and soft tissue engineering applications. It discusses agarose, alginate, cellulose, chitosan, collagen, decellularized extracellular matrix, dextran, fibrin, gelatin, gellan gum, hyaluronic acid, Matrigel, and silk. Multi-component bioinks are considered as a way to address the shortfalls of individual biomaterials. The mechanical, rheological, and cross-linking properties along with the cytocompatibility, cell viability, and printability of the bioinks are detailed as well. Future avenues for research into natural bioinks are then presented.
Collapse
Affiliation(s)
- Claire Benwood
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Josie Chrenek
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada; (J.C.); (H.R.); (K.T.)
| | - Rebecca L. Kirsch
- Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Nadia Z. Masri
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Hannah Richards
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada; (J.C.); (H.R.); (K.T.)
| | - Kyra Teetzen
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada; (J.C.); (H.R.); (K.T.)
| | - Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Biomedical Engineering Program, University of Victoria, Victoria, BC V8P 5C2, Canada; (J.C.); (H.R.); (K.T.)
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
| |
Collapse
|
107
|
Jamieson C, Keenan P, Kirkwood D, Oji S, Webster C, Russell KA, Koch TG. A Review of Recent Advances in 3D Bioprinting With an Eye on Future Regenerative Therapies in Veterinary Medicine. Front Vet Sci 2021; 7:584193. [PMID: 33665213 PMCID: PMC7921312 DOI: 10.3389/fvets.2020.584193] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/21/2020] [Indexed: 01/04/2023] Open
Abstract
3D bioprinting is a rapidly evolving industry that has been utilized for a variety of biomedical applications. It differs from traditional 3D printing in that it utilizes bioinks comprised of cells and other biomaterials to allow for the generation of complex functional tissues. Bioprinting involves computational modeling, bioink preparation, bioink deposition, and subsequent maturation of printed products; it is an intricate process where bioink composition, bioprinting approach, and bioprinter type must be considered during construct development. This technology has already found success in human studies, where a variety of functional tissues have been generated for both in vitro and in vivo applications. Although the main driving force behind innovation in 3D bioprinting has been utility in human medicine, recent efforts investigating its veterinary application have begun to emerge. To date, 3D bioprinting has been utilized to create bone, cardiovascular, cartilage, corneal and neural constructs in animal species. Furthermore, the use of animal-derived cells and various animal models in human research have provided additional information regarding its capacity for veterinary translation. While these studies have produced some promising results, technological limitations as well as ethical and regulatory challenges have impeded clinical acceptance. This article reviews the current understanding of 3D bioprinting technology and its recent advancements with a focus on recent successes and future translation in veterinary medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas G. Koch
- Reproductive Health and Biotechnology Lab, Department of Biomedical Science, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
108
|
Dobos A, Gantner F, Markovic M, Van Hoorick J, Tytgat L, Van Vlierberghe S, Ovsianikov A. On-chip high-definition bioprinting of microvascular structures. Biofabrication 2021; 13:015016. [PMID: 33586666 DOI: 10.1088/1758-5090/abb063] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
'Organ-on-chip' devices which integrate three-dimensional (3D) cell culture techniques with microfluidic approaches have the capacity to overcome the limitations of classical 2D platforms. Although several different strategies have been developed to improve the angiogenesis within hydrogels, one of the main challenges in tissue engineering remains the lack of vascularization in the fabricated 3D models. The present work focuses on the high-definition (HD) bioprinting of microvascular structures directly on-chip using two-photon polymerization (2PP). 2PP is a nonlinear process, where the near-infrared laser irradiation will only lead to the polymerization of a very small volume pixel (voxel), allowing the fabrication of channels in the microvascular range (10-30 µm in diameter). Additionally, 2PP not only enables the fabrication of sub-micrometer resolution scaffolds but also allows the direct embedding of cells within the produced structure. The accuracy of the 2PP printing parameters were optimized in order to achieve high-throughput and HD production of microfluidic vessel-on-chip platforms. The spherical aberrations stemming from the refractive index mismatch and the focusing depth inside the sample were simulated and the effect of the voxel compensation as well as different printing modes were demonstrated. Different layer spacings and their dependency on the applied laser power were compared both in terms of accuracy and required printing time resulting in a 10-fold decrease in structuring time while yielding well-defined channels of small diameters. Finally, the capacity of 2PP to create vascular structures within a microfluidic chip was tested with two different settings, by direct embedding of a co-culture of endothelial- and supporting cells during the printing process and by creating a supporting, cell-containing vascular scaffold barrier where the endothelial cell spheroids can be seeded afterwards. The functionality of the formed vessels was demonstrated with immunostaining of vascular endothelial cadherin (VE-Cadherin) endothelial adhesion molecules in both static and perfused culture.
Collapse
Affiliation(s)
- Agnes Dobos
- 3D Printing and Biofabrication Group, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria. Austrian Cluster for Tissue Regeneration (http://tissue-regeneration.at), Austria
| | | | | | | | | | | | | |
Collapse
|
109
|
Kronemberger GS, Carneiro FA, Rezende DF, Baptista LS. Spheroids and organoids as humanized 3D scaffold-free engineered tissues for SARS-CoV-2 viral infection and drug screening. Artif Organs 2021; 45:548-558. [PMID: 33264436 PMCID: PMC7753831 DOI: 10.1111/aor.13880] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022]
Abstract
The new coronavirus (2019‐nCoV) or the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) was officially declared by the World Health Organization (WHO) as a pandemic in March 2020. To date, there are no specific antiviral drugs proven to be effective in treating SARS‐CoV‐2, requiring joint efforts from different research fronts to discover the best route of treatment. The first decisions in drug discovery are based on 2D cell culture using high‐throughput screening. In this context, spheroids and organoids emerge as a reliable alternative. Both are scaffold‐free 3D engineered constructs that recapitulate key cellular and molecular events of tissue physiology. Different studies have already shown their advantages as a model for different infectious diseases, including SARS‐CoV‐2 and for drug screening. The use of these 3D engineered tissues as an in vitro model can fill the gap between 2D cell culture and in vivo preclinical assays (animal models) as they could recapitulate the entire viral life cycle. The main objective of this review is to understand spheroid and organoid biology, highlighting their advantages and disadvantages, and how these scaffold‐free engineered tissues can contribute to a better comprehension of viral infection by SARS‐CoV‐2 and to the development of in vitro high‐throughput models for drug screening.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Rio de Janeiro, Brazil.,Postgraduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, Brazil
| | - Fabiana A Carneiro
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Rio de Janeiro, Brazil
| | | | - Leandra S Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Rio de Janeiro, Brazil.,Postgraduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, Brazil
| |
Collapse
|
110
|
|
111
|
Xue C, Sutrisno L, Li M, Zhu W, Fei Y, Liu C, Wang X, Cai K, Hu Y, Luo Z. Implantable multifunctional black phosphorus nanoformulation-deposited biodegradable scaffold for combinational photothermal/ chemotherapy and wound healing. Biomaterials 2020; 269:120623. [PMID: 33388689 DOI: 10.1016/j.biomaterials.2020.120623] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/21/2022]
Abstract
Surgery is the mainstream treatment for melanoma, but its clinical implementation suffers from some major drawbacks including residual infiltrating melanoma cells at resection margins and severe tissue injury. In this study, a nanocomposite scaffold is developed for in-situ therapy after melanoma surgery as well as wound healing, which is fabricated by embedding photothermal-capable black phosphorus nanosheets (BPNSs) into bioresorbable Gelatin-PCL (GP) nanofibrous scaffold. GP scaffold is a clinically-tested biomaterial with temperature sensitivity and tissue-healing effect, while the BPNSs are loaded with the anticancer antibiotic of doxorubicin (DOX) and conjugated with NH2-PEG-FA for tumor-targeted delivery. The GP scaffold could undergo a sol-gel transition upon NIR irritation and release the BPNSs in situ. During this process, most of the BP-based nanoformulations were selectively internalized by the melanoma cells for the cooperative photothermal therapy and heat-triggerable DOX therapy, while some of the loaded DOX was released into the wound tissue to create a tumor-suppressive microenvironment. Moreover, BPNSs could be gradually degraded to phosphates/phosphonates and thus enhance tissue repair by activating the ERK1/2 and PI3K/Akt pathway. Meanwhile, the detached DOX molecules would also enter the wound tissues for continuous melanoma inhibition. Considering the anti-melanoma and wound healing effect of this composite scaffold, it may offer a facile strategy for the wound treatment after melanoma surgery.
Collapse
Affiliation(s)
- Chencheng Xue
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Linawati Sutrisno
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Wei Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Changhuang Liu
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Xuan Wang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
112
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [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: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
113
|
Kim SJ, Kim EM, Yamamoto M, Park H, Shin H. Engineering Multi-Cellular Spheroids for Tissue Engineering and Regenerative Medicine. Adv Healthc Mater 2020; 9:e2000608. [PMID: 32734719 DOI: 10.1002/adhm.202000608] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Multi-cellular spheroids are formed as a 3D structure with dense cell-cell/cell-extracellular matrix interactions, and thus, have been widely utilized as implantable therapeutics and various ex vivo tissue models in tissue engineering. In principle, spheroid culture methods maximize cell-cell cohesion and induce spontaneous cellular assembly while minimizing cellular interactions with substrates by using physical forces such as gravitational or centrifugal forces, protein-repellant biomaterials, and micro-structured surfaces. In addition, biofunctional materials including magnetic nanoparticles, polymer microspheres, and nanofiber particles are combined with cells to harvest composite spheroids, to accelerate spheroid formation, to increase the mechanical properties and viability of spheroids, and to direct differentiation of stem cells into desirable cell types. Biocompatible hydrogels are developed to produce microgels for the fabrication of size-controlled spheroids with high efficiency. Recently, spheroids have been further engineered to fabricate structurally and functionally reliable in vitro artificial 3D tissues of the desired shape with enhanced specific biological functions. This paper reviews the overall characteristics of spheroids and general/advanced spheroid culture techniques. Significant roles of functional biomaterials in advanced spheroid engineering with emphasis on the use of spheroids in the reconstruction of artificial 3D tissue for tissue engineering are also thoroughly discussed.
Collapse
Affiliation(s)
- Se-Jeong Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Eun Mi Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Masaya Yamamoto
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
- Biomedical Engineering for Diagnosis and Treatment, Graduate School of Biomedical Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Hansoo Park
- School of Integrative Engineering, College of Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Institute of Nano Science & Technology (INST), Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| |
Collapse
|
114
|
Maina RM, Barahona MJ, Geibel P, Lysyy T, Finotti M, Isaji T, Wengerter B, Mentone S, Dardik A, Geibel JP. Hydrogel-based 3D bioprints repair rat small intestine injuries and integrate into native intestinal tissue. J Tissue Eng Regen Med 2020; 15:129-138. [PMID: 33197151 DOI: 10.1002/term.3157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 10/18/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022]
Abstract
3D Printing has become a mainstay of industry, with several applications in the medical field. One area that could benefit from 3D printing is intestinal failure due to injury or genetic malformations. We bioprinted cylindrical tubes from rat vascular cells that were sized to form biopatches. 2 mm enterotomies were made in the small intestine of male Sprague-Dawley rats, and sealed with biopatches. These intestinal segments were connected to an ex vivo perfusion device that provided independent extraluminal and intraluminal perfusion. The fluorescence signal of fluorescein isothiocyanate (FITC)-inulin in the intraluminal perfusate, a non-absorbable fluorescent marker of intestinal integrity, was measured every 15 min over 90 min, and used to assess the integrity of the segments under both continuous perfusion and alternate-flow perfusion. Enterotomies were made an inch away from the ileocecal junction in male Wistar rats and sealed with biopatches. The animals were monitored daily and euthanized at post-operative days 7, 14, 21, and 30. Blinded histopathological analysis was conducted to compare the patch segments to native intestine. Biopatch-sealed intestinal segments withstood both continuous and pulsatile flow rates without leakage of FITC-inulin above the control baseline. 21 of 26 animals survived with normal activity, weight gain, and stool output. Histopathology of the explanted segments showed progressive villi and crypt formation over the enterotomies, with complete restoration of the epithelium by 30 days. This study presents a novel application of 3D bioprinting to develop a universal repair patch that can seal lesions in vivo, and fully integrate into the native intestine.
Collapse
Affiliation(s)
- Renee M Maina
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Maria J Barahona
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Peter Geibel
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Taras Lysyy
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michele Finotti
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA.,Transplantation & Hepatobiliary Surgery, University of Padova, Padova, Italy
| | - Toshihiko Isaji
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Brian Wengerter
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - SueAnn Mentone
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Alan Dardik
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - John P Geibel
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
115
|
Fröhlich E. Issues with Cancer Spheroid Models in Therapeutic Drug Screening. Curr Pharm Des 2020; 26:2137-2148. [PMID: 32067603 DOI: 10.2174/1381612826666200218094200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/17/2020] [Indexed: 12/31/2022]
Abstract
In vitro screening for anti-cancer agents currently uses mainly cell lines in 2D culture. It is generally assumed that 3D culture, namely spheroids, represents physiologically more relevant models for tumors. Unfortunately, drug testing in spheroids is not as easy and reproducible as in 2D culture because there are factors that limit the universal use of spheroids as screening platforms. Technical problems in the generation of uniform spheroids, cell/tumor-specific differences in the ability to form spheroids, and more complex readout parameters are the main reasons for differences between spheroid data. The review discusses requirements for cancer spheroids to be representative models, suitable methodologies to generate spheroids for the screening and readout parameters for the evaluation of anti-cancer agents.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
| |
Collapse
|
116
|
Santos LF, Sofia Silva A, Mano JF. Complex-shaped magnetic 3D cell-based structures for tissue engineering. Acta Biomater 2020; 118:18-31. [PMID: 33039596 DOI: 10.1016/j.actbio.2020.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022]
Abstract
The use of cells as building blocks for tissue engineering purposes has been a matter of research in the recent years. Still, the fabrication of complex-shaped 3D-like constructs using living-based materials is hampered through the difficulty in recapitulating the mechanical properties of the native tissues. In an attempt to develop robust tissue-like constructs, it is herein proposed the fabrication of complex-shaped magnetic cell sheets (CSs) with improved mechanical properties for bone TE. Hence, magnetic CSs with versatile shapes and enhanced mechanical performance are fabricated using a pre-osteoblast cell line (MC3T3-E1) through an universal approach that relies on the design of the substrate, cell density and magnetic force. Results show that such magnetic CSs exhibit a Young's modulus similar to those encountered in the soft tissues. The construction of stratified CSs is also explored using MC3T3-E1 and adipose-derived stromal cells (ASCs). The role of the pre-osteoblast cell line on ASCs osteogenesis is herein investigated for the first time in layered scaffold-free structures. After 21 days, the level of osteogenic markers in the heterotypic CS (MC3T3-E1:ASCs) is significantly higher than in the homotypic one (ASCs:ASCs), even in the absence of osteogenic differentiation factors. These evidences open new prospects for the creation of mechanically robust, complex, higher-ordered and completely functional 3D cell-based materials that better resemble the native environment of in vivo tissues.
Collapse
|
117
|
Lee SW, Jung DJ, Jeong GS. Gaining New Biological and Therapeutic Applications into the Liver with 3D In Vitro Liver Models. Tissue Eng Regen Med 2020; 17:731-745. [PMID: 32207030 PMCID: PMC7710770 DOI: 10.1007/s13770-020-00245-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cell cultures with architectural and biomechanical properties similar to those of natural tissue have been the focus for generating liver tissue. Microarchitectural organization is believed to be crucial to hepatic function, and 3D cell culture technologies have enabled the construction of tissue-like microenvironments, thereby leading to remarkable progress in vitro models of human tissue and organs. Recently, to recapitulate the 3D architecture of tissues, spheroids and organoids have become widely accepted as new practical tools for 3D organ modeling. Moreover, the combination of bioengineering approach offers the promise to more accurately model the tissue microenvironment of human organs. Indeed, the employment of sophisticated bioengineered liver models show long-term viability and functional enhancements in biochemical parameters and disease-orient outcome. RESULTS Various 3D in vitro liver models have been proposed as a new generation of liver medicine. Likewise, new biomedical engineering approaches and platforms are available to more accurately replicate the in vivo 3D microarchitectures and functions of living organs. This review aims to highlight the recent 3D in vitro liver model systems, including micropatterning, spheroids, and organoids that are either scaffold-based or scaffold-free systems. Finally, we discuss a number of challenges that will need to be addressed moving forward in the field of liver tissue engineering for biomedical applications. CONCLUSION The ongoing development of biomedical engineering holds great promise for generating a 3D biomimetic liver model that recapitulates the physiological and pathological properties of the liver and has biomedical applications.
Collapse
Affiliation(s)
- Sang Woo Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Da Jung Jung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
- Department of Convergence Medicine, University of Ulsan College of Medicine, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
118
|
Sarigil O, Anil-Inevi M, Firatligil-Yildirir B, Unal YC, Yalcin-Ozuysal O, Mese G, Tekin HC, Ozcivici E. Scaffold-free biofabrication of adipocyte structures with magnetic levitation. Biotechnol Bioeng 2020; 118:1127-1140. [PMID: 33205833 DOI: 10.1002/bit.27631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/27/2020] [Accepted: 11/15/2020] [Indexed: 12/16/2022]
Abstract
Tissue engineering research aims to repair the form and/or function of impaired tissues. Tissue engineering studies mostly rely on scaffold-based techniques. However, these techniques have certain challenges, such as the selection of proper scaffold material, including mechanical properties, sterilization, and fabrication processes. As an alternative, we propose a novel scaffold-free adipose tissue biofabrication technique based on magnetic levitation. In this study, a label-free magnetic levitation technique was used to form three-dimensional (3D) scaffold-free adipocyte structures with various fabrication strategies in a microcapillary-based setup. Adipogenic-differentiated 7F2 cells and growth D1 ORL UVA stem cells were used as model cells. The morphological properties of the 3D structures of single and cocultured cells were analyzed. The developed procedure leads to the formation of different patterns of single and cocultured adipocytes without a scaffold. Our results indicated that adipocytes formed loose structures while growth cells were tightly packed during 3D culture in the magnetic levitation platform. This system has potential for ex vivo modeling of adipose tissue for drug testing and transplantation applications for cell therapy in soft tissue damage. Also, it will be possible to extend this technique to other cell and tissue types.
Collapse
Affiliation(s)
- Oyku Sarigil
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Muge Anil-Inevi
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | | | - Yagmur Ceren Unal
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Ozden Yalcin-Ozuysal
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Gulistan Mese
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - H Cumhur Tekin
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Engin Ozcivici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| |
Collapse
|
119
|
Nosrati H, Alizadeh Z, Nosrati A, Ashrafi-Dehkordi K, Banitalebi-Dehkordi M, Sanami S, Khodaei M. Stem cell-based therapeutic strategies for corneal epithelium regeneration. Tissue Cell 2020; 68:101470. [PMID: 33248403 DOI: 10.1016/j.tice.2020.101470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Any significant loss of vision or blindness caused by corneal damages is referred to as corneal blindness. Corneal blindness is the fourth most common cause of blindness worldwide, representing more than 5% of the total blind population. Currently, corneal transplantation is used to treat many corneal diseases. In some cases, implantation of artificial cornea (keratoprosthesis) is suggested after a patient has had a donor corneal transplant failure. The shortage of donors and the side effects of keratoprosthesis are limiting these approaches. Recently, researchers have been actively pursuing new approaches for corneal regeneration because of these limitations. Nowadays, tissue engineering of different corneal layers (epithelium, stroma, endothelium, or full thickness tissue) is a promising approach that has attracted a great deal of interest from researchers and focuses on regenerative strategies using different cell sources and biomaterials. Various sources of corneal and non-corneal stem cells have shown significant advantages for corneal epithelium regeneration applications. Pluripotent stem cells (embryonic stem cells and iPS cells), epithelial stem cells (derived from oral mucus, amniotic membrane, epidermis and hair follicle), mesenchymal stem cells (bone marrow, adipose-derived, amniotic membrane, placenta, umbilical cord), and neural crest origin stem cells (dental pulp stem cells) are the most promising sources in this regard. These cells could also be used in combination with natural or synthetic scaffolds to improve the efficacy of the therapeutic approach. As the ocular surface is exposed to external damage, the number of studies on regeneration of the corneal epithelium is rising. In this paper, we reviewed the stem cell-based strategies for corneal epithelium regeneration.
Collapse
Affiliation(s)
- Hamed Nosrati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Zohreh Alizadeh
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Nosrati
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Korosh Ashrafi-Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehdi Banitalebi-Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Sanami
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Khodaei
- Department of Materials Science and Engineering, Golpayegan University of Technology, Golpayegan, Iran
| |
Collapse
|
120
|
Nosrati H, Abpeikar Z, Mahmoudian ZG, Zafari M, Majidi J, Alizadeh A, Moradi L, Asadpour S. Corneal epithelium tissue engineering: recent advances in regeneration and replacement of corneal surface. Regen Med 2020; 15:2029-2044. [PMID: 33169642 DOI: 10.2217/rme-2019-0055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Currently, many corneal diseases are treated by corneal transplantation, artificial corneal implantation or, in severe cases, keratoprosthesis. Owing to the shortage of cornea donors and the risks involved with artificial corneal implants, such as infection transmission, researchers continually seek new approaches for corneal regeneration. Corneal tissue engineering is a promising approach that has attracted much attention from researchers and is focused on regenerative strategies using various biomaterials in combination with different cell types. These constructs should have the ability to mimic the native tissue microenvironment and present suitable optical, mechanical and biological properties. In this article, we review studies that have focused on the current clinical techniques for corneal replacement. We also describe tissue-engineering and cell-based approaches for corneal regeneration.
Collapse
Affiliation(s)
- Hamed Nosrati
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Gholami Mahmoudian
- Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahdi Zafari
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Jafar Majidi
- Cellular & Molecular Research Center, Basic Health Science Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering & Applied Cell Sciences, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Lida Moradi
- The Ronald O Perelman Department of Dermatology, New York University, School of Medicine, New York, NY 10016, USA.,Department of Cell Biology, New York University, School of Medicine, New York, NY, 10016 USA
| | - Shiva Asadpour
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Cellular & Molecular Research Center, Basic Health Science Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
121
|
Pan Y, Xie Z, Cen S, Li M, Liu W, Tang S, Ye G, Li J, Zheng G, Li Z, Yu W, Wang P, Wu Y, Shen H. Long noncoding RNA repressor of adipogenesis negatively regulates the adipogenic differentiation of mesenchymal stem cells through the hnRNP A1-PTX3-ERK axis. Clin Transl Med 2020; 10:e227. [PMID: 33252864 PMCID: PMC7648959 DOI: 10.1002/ctm2.227] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are pluripotent stem cells that can differentiate via osteogenesis and adipogenesis. The mechanism underlying MSC lineage commitment still remains incompletely elucidated. Understanding the regulatory mechanism of MSC differentiation will help researchers induce MSCs toward specific lineages for clinical use. In this research, we intended to figure out the long noncoding RNA (lncRNA) that plays a central role in MSC fate determination and explore its application value in tissue engineering. METHODS The expression pattern of lncRNAs during MSC osteogenesis/adipogenesis was detected by microarray and qRT-PCR. Lentivirus and siRNAs were constructed to regulate the expression of lncRNA repressor of adipogenesis (ROA). MSC osteogenesis/adipogenesis was evaluated by western blot and alizarin red/oil red staining. An adipokine array was used to select the paracrine/autocrine factor PTX3, followed by RNA interference or recombinant human protein stimulation to confirm its function. The activation of signaling pathways was also detected by western blot, and a small molecule inhibitor, SCH772984, was used to inhibit the activation of the ERK pathway. The interaction between ROA and hnRNP A1 was detected by RNA pull-down and RIP assays. Luciferase reporter and chromatin immunoprecipitation assays were used to confirm the binding of hnRNP A1 to the PTX3 promotor. Additionally, an in vivo adipogenesis experiment was conducted to evaluate the regulatory value of ROA in tissue engineering. RESULTS In this study, we demonstrated that MSC adipogenesis is regulated by lncRNA ROA both in vitro and in vivo. Mechanistically, ROA inhibits MSC adipogenesis by downregulating the expression of the key autocrine/paracrine factor PTX3 and the downstream ERK pathway. This downregulation was achieved through transcription inhibition by impeding hnRNP A1 from binding to the promoter of PTX3. CONCLUSIONS ROA negatively regulates MSC adipogenesis through the hnRNP A1-PTX3-ERK axis. ROA may be an effective target for modulating MSCs in tissue engineering.
Collapse
Affiliation(s)
- Yiqian Pan
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhongyu Xie
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Shuizhong Cen
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of OrthopedicsZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ming Li
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Wenjie Liu
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Su'an Tang
- Clinical Research CenterZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guiwen Ye
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jinteng Li
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Guan Zheng
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Zhaofeng Li
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Wenhui Yu
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Peng Wang
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Yanfeng Wu
- Center for BiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Huiyong Shen
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
122
|
Perez JE, Nagle I, Wilhelm C. Magnetic molding of tumor spheroids: emerging model for cancer screening. Biofabrication 2020; 13. [PMID: 33126227 DOI: 10.1088/1758-5090/abc670] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023]
Abstract
Three-dimensional tissue culture, and particularly spheroid models, have recently been recognized as highly relevant in drug screening, toxicity assessment and tissue engineering due to their superior complexity and heterogeneity akin to the in vivo microenvironment. However, limitations in size control, shape reproducibility and long maturation times hinder their full applicability. Here, we report a spheroid formation technique based on the magnetic aggregation of cells with internalized magnetic nanoparticles. The method yields magnetic spheroids with high sphericity and allows fine-tuning the final spheroid diameter. Moreover, cohesive spheroids can be obtained in less than 24 hours. We show the proof of concept of the method using the CT26 murine colon carcinoma cell line and how different cell proliferation and invasion potentials can be attained by varying the spheroid size. Additionally, we show how the spheroid maturation impacts cell invasion and doxorubicin penetrability, highlighting the importance of this parameter in drug screening and therapeutic applications. Finally, we demonstrate the capability of the method to allow the measurement of the spheroid surface tension, a relevant output parameter in the context of cancer cell invasion and metastasis. The method can accommodate other cell lines able to be magnetically labeled, as we demonstrate using the U-87 MG human glioblastoma cell line, and shows promise in the therapeutic screening at early time points of tissue formation, as well as in studies of drug and nanoparticle tumor penetration.
Collapse
Affiliation(s)
- Jose Efrain Perez
- Laboratoire Matiere et Systemes Complexes UMR CNRS 7057, University of Paris, Paris, FRANCE
| | - Irène Nagle
- Laboratoire Matiere et Systemes Complexes UMR CNRS 7057, University of Paris, Paris, FRANCE
| | - Claire Wilhelm
- Laboratoire Matiere et Systemes Complexes UMR CNRS 7057, University of Paris, Batiment Condorcet, 10 rue Alice Domon et Leonie Duquet, 75025 Paris Cedax 13, Paris, 75013, FRANCE
| |
Collapse
|
123
|
Kim J, Kong JS, Han W, Kim BS, Cho DW. 3D Cell Printing of Tissue/Organ-Mimicking Constructs for Therapeutic and Drug Testing Applications. Int J Mol Sci 2020; 21:E7757. [PMID: 33092184 PMCID: PMC7589604 DOI: 10.3390/ijms21207757] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The development of artificial tissue/organs with the functional maturity of their native equivalents is one of the long-awaited panaceas for the medical and pharmaceutical industries. Advanced 3D cell-printing technology and various functional bioinks are promising technologies in the field of tissue engineering that have enabled the fabrication of complex 3D living tissue/organs. Various requirements for these tissues, including a complex and large-volume structure, tissue-specific microenvironments, and functional vasculatures, have been addressed to develop engineered tissue/organs with native relevance. Functional tissue/organ constructs have been developed that satisfy such criteria and may facilitate both in vivo replenishment of damaged tissue and the development of reliable in vitro testing platforms for drug development. This review describes key developments in technologies and materials for engineering 3D cell-printed constructs for therapeutic and drug testing applications.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Wonil Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
124
|
Ramesh S, Kovelakuntla V, Meyer AS, Rivero IV. Three-dimensional printing of stimuli-responsive hydrogel with antibacterial activity. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
125
|
Abdollahiyan P, Oroojalian F, Mokhtarzadeh A, Guardia M. Hydrogel‐Based 3D Bioprinting for Bone and Cartilage Tissue Engineering. Biotechnol J 2020; 15:e2000095. [DOI: 10.1002/biot.202000095] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/22/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Parinaz Abdollahiyan
- Immunology Research Center Tabriz University of Medical Sciences Tabriz 5166614731 Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies School of Medicine North Khorasan University of Medical Sciences Bojnurd 7487794149 Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center Tabriz University of Medical Sciences Tabriz 5166614731 Iran
| | - Miguel Guardia
- Department of Analytical Chemistry University of Valencia Dr. Moliner 50 Burjassot Valencia 46100 Spain
| |
Collapse
|
126
|
Bédard P, Gauvin S, Ferland K, Caneparo C, Pellerin È, Chabaud S, Bolduc S. Innovative Human Three-Dimensional Tissue-Engineered Models as an Alternative to Animal Testing. Bioengineering (Basel) 2020; 7:E115. [PMID: 32957528 PMCID: PMC7552665 DOI: 10.3390/bioengineering7030115] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Animal testing has long been used in science to study complex biological phenomena that cannot be investigated using two-dimensional cell cultures in plastic dishes. With time, it appeared that more differences could exist between animal models and even more when translated to human patients. Innovative models became essential to develop more accurate knowledge. Tissue engineering provides some of those models, but it mostly relies on the use of prefabricated scaffolds on which cells are seeded. The self-assembly protocol has recently produced organ-specific human-derived three-dimensional models without the need for exogenous material. This strategy will help to achieve the 3R principles.
Collapse
Affiliation(s)
- Patrick Bédard
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Sara Gauvin
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Karel Ferland
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Ève Pellerin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
127
|
Bansal SA, Kumar V, Karimi J, Singh AP, Kumar S. Role of gold nanoparticles in advanced biomedical applications. NANOSCALE ADVANCES 2020; 2:3764-3787. [PMID: 36132791 PMCID: PMC9419294 DOI: 10.1039/d0na00472c] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/14/2020] [Indexed: 05/20/2023]
Abstract
Gold nanoparticles (GNPs) have generated keen interest among researchers in recent years due to their excellent physicochemical properties. In general, GNPs are biocompatible, amenable to desired functionalization, non-corroding, and exhibit size and shape dependent optical and electronic properties. These excellent properties of GNPs exhibit their tremendous potential for use in diverse biomedical applications. Herein, we have evaluated the recent advancements of GNPs to highlight their exceptional potential in the biomedical field. Special focus has been given to emerging biomedical applications including bio-imaging, site specific drug/gene delivery, nano-sensing, diagnostics, photon induced therapeutics, and theranostics. We have also elaborated on the basics, presented a historical preview, and discussed the synthesis strategies, functionalization methods, stabilization techniques, and key properties of GNPs. Lastly, we have concluded this article with key findings and unaddressed challenges. Overall, this review is a complete package to understand the importance and achievements of GNPs in the biomedical field.
Collapse
Affiliation(s)
- Suneev Anil Bansal
- Department of Mechanical Engineering, University Institute of Engineering and Technology (UIET), Panjab University Chandigarh India 160014
- Department of Mechanical Engineering, MAIT, Maharaja Agrasen University HP India 174103
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI) S. A. S. Nagar Punjab 140306 India
| | - Javad Karimi
- Department of Biology, Faculty of Sciences, Shiraz University Shiraz 71454 Iran
| | - Amrinder Pal Singh
- Department of Mechanical Engineering, University Institute of Engineering and Technology (UIET), Panjab University Chandigarh India 160014
| | - Suresh Kumar
- Department of Applied Science, University Institute of Engineering and Technology (UIET), Panjab University Chandigarh India 160014
| |
Collapse
|
128
|
|
129
|
Dissanayaka WL, Zhang C. Scaffold-based and Scaffold-free Strategies in Dental Pulp Regeneration. J Endod 2020; 46:S81-S89. [DOI: 10.1016/j.joen.2020.06.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
130
|
Rasoulinejad S, Mueller M, Nzigou Mombo B, Wegner SV. Orthogonal Blue and Red Light Controlled Cell-Cell Adhesions Enable Sorting-out in Multicellular Structures. ACS Synth Biol 2020; 9:2076-2086. [PMID: 32610009 PMCID: PMC7757848 DOI: 10.1021/acssynbio.0c00150] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
![]()
The self-assembly of different cell
types into multicellular structures
and their organization into spatiotemporally controlled patterns are
both challenging and extremely powerful to understand how cells function
within tissues and for bottom-up tissue engineering. Here, we not
only independently control the self-assembly of two cell types into
multicellular architectures with blue and red light, but also achieve
their self-sorting into distinct assemblies. This required developing
two cell types that form selective and homophilic cell–cell
interactions either under blue or red light using photoswitchable
proteins as artificial adhesion molecules. The interactions were individually
triggerable with different colors of light, reversible in the dark,
and provide noninvasive and temporal control over the cell–cell
adhesions. In mixtures of the two cells, each cell type self-assembled
independently upon orthogonal photoactivation, and cells sorted out
into separate assemblies based on specific self-recognition. These
self-sorted multicellular architectures provide us with a powerful
tool for producing tissue-like structures from multiple cell types
and investigate principles that govern them.
Collapse
Affiliation(s)
- Samaneh Rasoulinejad
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - Marc Mueller
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - Brice Nzigou Mombo
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster Waldeyerstrasse 15, Münster, 48149, Germany
| | - Seraphine V. Wegner
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster Waldeyerstrasse 15, Münster, 48149, Germany
| |
Collapse
|
131
|
Chitosan-Hydrogel Polymeric Scaffold Acts as an Independent Primary Inducer of Osteogenic Differentiation in Human Mesenchymal Stromal Cells. MATERIALS 2020; 13:ma13163546. [PMID: 32796668 PMCID: PMC7475832 DOI: 10.3390/ma13163546] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Regenerative medicine aims to restore damaged tissues and mainly takes advantage of human mesenchymal stromal cells (hMSCs), either alone or combined with three-dimensional scaffolds. The scaffold is generally considered a support, and its contribution to hMSC proliferation and differentiation is unknown or poorly investigated. The aim of this study was to evaluate the capability of an innovative three-dimensional gelatin–chitosan hybrid hydrogel scaffold (HC) to activate the osteogenic differentiation process in hMSCs. We seeded hMSCs from adipose tissue (AT-hMSCs) and bone marrow (BM-hMSCs) in highly performing HC of varying chitosan content in the presence of growing medium (GM) or osteogenic medium (OM) combined with Fetal Bovine Serum (FBS) or human platelet lysate (hPL). We primarily evaluated the viability and the proliferation of AT-hMSCs and BM-hMSCs under different conditions. Then, in order to analyse the activation of osteogenic differentiation, the osteopontin (OPN) transcript was absolutely quantified at day 21 by digital PCR. OPN was expressed under all conditions, in both BM-hMSCs and AT-hMSCs. Cells seeded in HC cultured with OM+hPL presented the highest OPN transcript levels, as expected. Interestingly, both BM-hMSCs and AT-hMSCs cultured with GM+FBS expressed OPN. In particular, BM-hMSCs cultured with GM+FBS expressed more OPN than those cultured with GM+hPL and OM+FBS; AT-hMSCs cultured with GM+FBS presented a lower expression of OPN when compared with those cultured with GM+hPL, but no significant difference was detected when compared with AT-hMSCs cultured with OM+FBS. No OPN expression was detected in negative controls. These results show the capability of HC to primarily and independently activate osteogenic differentiation pathways in hMCSs. Therefore, these scaffolds may be considered no more as a simple support, rather than active players in the differentiative and regenerative process.
Collapse
|
132
|
Gupta S, Sharma A, Vasantha Kumar J, Sharma V, Gupta PK, Verma RS. Meniscal tissue engineering via 3D printed PLA monolith with carbohydrate based self-healing interpenetrating network hydrogel. Int J Biol Macromol 2020; 162:1358-1371. [PMID: 32777410 DOI: 10.1016/j.ijbiomac.2020.07.238] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
Failure of bioengineered meniscus implant after transplantation is a major concern owing to mechanical failure, lack of chondrogenic capability and patient specific design. In this article, we have, for the first time, fabricated a 3D printed scaffold with carbohydrate based self-healing interpenetrating network (IPN) hydrogels-based monolith construct for load bearing meniscus tissue. 3D printed PLA scaffold was surface functionalized and embedded with self-healing IPN hydrogel for interfacial bonding further characterized by micro CT. Using collagen (C), alginate (A) and oxidized alginate (ADA), we developed self-healing IPN hydrogels with dual crosslinking (Ca2+ based ionic crosslinking and Schiff base (A-A, A-ADA)) capability and studied their physicochemical properties. Further, we studied human stem cells behaviour and chondrogenic differentiation potential within these IPN hydrogels. In-vivo heterotopic implantation confirmed biocompatibility of the monolith showing the feasibility of using carbohydrate based IPN hydrogel embedded in 3D printed scaffold for meniscal tissue development.
Collapse
Affiliation(s)
- Santosh Gupta
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Akriti Sharma
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - J Vasantha Kumar
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Vineeta Sharma
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Piyush Kumar Gupta
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| |
Collapse
|
133
|
Vyas C, Mishbak H, Cooper G, Peach C, Pereira RF, Bartolo P. Biological perspectives and current biofabrication strategies in osteochondral tissue engineering. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s40898-020-00008-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractArticular cartilage and the underlying subchondral bone are crucial in human movement and when damaged through disease or trauma impacts severely on quality of life. Cartilage has a limited regenerative capacity due to its avascular composition and current therapeutic interventions have limited efficacy. With a rapidly ageing population globally, the numbers of patients requiring therapy for osteochondral disorders is rising, leading to increasing pressures on healthcare systems. Research into novel therapies using tissue engineering has become a priority. However, rational design of biomimetic and clinically effective tissue constructs requires basic understanding of osteochondral biological composition, structure, and mechanical properties. Furthermore, consideration of material design, scaffold architecture, and biofabrication strategies, is needed to assist in the development of tissue engineering therapies enabling successful translation into the clinical arena. This review provides a starting point for any researcher investigating tissue engineering for osteochondral applications. An overview of biological properties of osteochondral tissue, current clinical practices, the role of tissue engineering and biofabrication, and key challenges associated with new treatments is provided. Developing precisely engineered tissue constructs with mechanical and phenotypic stability is the goal. Future work should focus on multi-stimulatory environments, long-term studies to determine phenotypic alterations and tissue formation, and the development of novel bioreactor systems that can more accurately resemble the in vivo environment.
Collapse
|
134
|
Sha'ban M, Ahmad Radzi MA. Scaffolds for Cartilage Regeneration: To Use or Not to Use? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1249:97-114. [PMID: 32602093 DOI: 10.1007/978-981-15-3258-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Joint cartilage has been a significant focus on the field of tissue engineering and regenerative medicine (TERM) since its inception in the 1980s. Represented by only one cell type, cartilage has been a simple tissue that is thought to be straightforward to deal with. After three decades, engineering cartilage has proven to be anything but easy. With the demographic shift in the distribution of world population towards ageing, it is expected that there is a growing need for more effective options for joint restoration and repair. Despite the increasing understanding of the factors governing cartilage development, there is still a lot to do to bridge the gap from bench to bedside. Dedicated methods to regenerate reliable articular cartilage that would be equivalent to the original tissue are still lacking. The use of cells, scaffolds and signalling factors has always been central to the TERM. However, without denying the importance of cells and signalling factors, the question posed in this chapter is whether the answer would come from the methods to use or not to use scaffold for cartilage TERM. This paper presents some efforts in TERM area and proposes a solution that will transpire from the ongoing attempts to understand certain aspects of cartilage development, degeneration and regeneration. While an ideal formulation for cartilage regeneration has yet to be resolved, it is felt that scaffold is still needed for cartilage TERM for years to come.
Collapse
Affiliation(s)
- Munirah Sha'ban
- Department of Physical Rehabilitation Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia.
| | - Muhammad Aa'zamuddin Ahmad Radzi
- Department of Biomedical Science, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| |
Collapse
|
135
|
Parfenov VA, Khesuani YD, Petrov SV, Karalkin PA, Koudan EV, Nezhurina EK, Pereira FDAS, Krokhmal AA, Gryadunova AA, Bulanova EA, Vakhrushev IV, Babichenko II, Kasyanov V, Petrov OF, Vasiliev MM, Brakke K, Belousov SI, Grigoriev TE, Osidak EO, Rossiyskaya EI, Buravkova LB, Kononenko OD, Demirci U, Mironov VA. Magnetic levitational bioassembly of 3D tissue construct in space. SCIENCE ADVANCES 2020; 6:eaba4174. [PMID: 32743068 PMCID: PMC7363443 DOI: 10.1126/sciadv.aba4174] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/04/2020] [Indexed: 05/02/2023]
Abstract
Magnetic levitational bioassembly of three-dimensional (3D) tissue constructs represents a rapidly emerging scaffold- and label-free approach and alternative conceptual advance in tissue engineering. The magnetic bioassembler has been designed, developed, and certified for life space research. To the best of our knowledge, 3D tissue constructs have been biofabricated for the first time in space under microgravity from tissue spheroids consisting of human chondrocytes. Bioassembly and sequential tissue spheroid fusion presented a good agreement with developed predictive mathematical models and computer simulations. Tissue constructs demonstrated good viability and advanced stages of tissue spheroid fusion process. Thus, our data strongly suggest that scaffold-free formative biofabrication using magnetic fields is a feasible alternative to traditional scaffold-based approaches, hinting a new perspective avenue of research that could significantly advance tissue engineering. Magnetic levitational bioassembly in space can also advance space life science and space regenerative medicine.
Collapse
Affiliation(s)
- Vladislav A. Parfenov
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
- A.A. Baikov Institute of Metallurgy and Material Science, Russian Academy of Sciences, Moscow, Russia
- Corresponding author. (V.A.P.); (V.A.M.); (U.D.)
| | - Yusef D. Khesuani
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Stanislav V. Petrov
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Pavel A. Karalkin
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, Moscow, Russia
| | - Elizaveta V. Koudan
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Elizaveta K. Nezhurina
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, Moscow, Russia
| | | | - Alisa A. Krokhmal
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Anna A. Gryadunova
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Elena A. Bulanova
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Igor V. Vakhrushev
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
| | - Igor I. Babichenko
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | | | - Oleg F. Petrov
- Joint Institute for High Temperatures, Russian Academy of Sciences, Moscow, Russia
| | - Mikhail M. Vasiliev
- Joint Institute for High Temperatures, Russian Academy of Sciences, Moscow, Russia
| | - Kenn Brakke
- Susquehanna University, Selinsgrove, PA, USA
| | | | | | | | | | | | - Oleg D. Kononenko
- Yu.A. Gagarin Research & Test Cosmonaut Training Center, Star City, Moscow Region, Russia
| | - Utkan Demirci
- Canary Center for Early Cancer Detection, Department of Radiology, Stanford University, Palo Alto, CA, USA
- Corresponding author. (V.A.P.); (V.A.M.); (U.D.)
| | - Vladimir A. Mironov
- Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Corresponding author. (V.A.P.); (V.A.M.); (U.D.)
| |
Collapse
|
136
|
Sun H, Jia Y, Dong H, Dong D, Zheng J. Combining additive manufacturing with microfluidics: an emerging method for developing novel organs-on-chips. Curr Opin Chem Eng 2020. [DOI: 10.1016/j.coche.2019.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
137
|
Dalton PD, Woodfield TBF, Mironov V, Groll J. Advances in Hybrid Fabrication toward Hierarchical Tissue Constructs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902953. [PMID: 32537395 PMCID: PMC7284200 DOI: 10.1002/advs.201902953] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/17/2020] [Indexed: 05/05/2023]
Abstract
The diversity of manufacturing processes used to fabricate 3D implants, scaffolds, and tissue constructs is continuously increasing. This growing number of different applicable fabrication technologies include electrospinning, melt electrowriting, volumetric-, extrusion-, and laser-based bioprinting, the Kenzan method, and magnetic and acoustic levitational bioassembly, to name a few. Each of these fabrication technologies feature specific advantages and limitations, so that a combination of different approaches opens new and otherwise unreachable opportunities for the fabrication of hierarchical cell-material constructs. Ongoing challenges such as vascularization, limited volume, and repeatability of tissue constructs at the resolution required to mimic natural tissue is most likely greater than what one manufacturing technology can overcome. Therefore, the combination of at least two different manufacturing technologies is seen as a clear and necessary emerging trend, especially within biofabrication. This hybrid approach allows more complex mechanics and discrete biomimetic structures to address mechanotransduction and chemotactic/haptotactic cues. Pioneering milestone papers in hybrid fabrication for biomedical purposes are presented and recent trends toward future manufacturing platforms are analyzed.
Collapse
Affiliation(s)
- Paul D. Dalton
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity of WürzburgWürzburg97070Germany
| | - Tim B. F. Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of Otago ChristchurchChristchurch8011New Zealand
- New Zealand Medical Technologies Centre of Research Excellence (MedTech CoRE)Auckland0600‐2699New Zealand
| | - Vladimir Mironov
- 3D Bioprinting SolutionsMoscow115409Russia
- Institute for Regenerative MedicineSechenov Medical UniversityMoscow119992Russia
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity of WürzburgWürzburg97070Germany
| |
Collapse
|
138
|
Ntege EH, Sunami H, Shimizu Y. Advances in regenerative therapy: A review of the literature and future directions. Regen Ther 2020; 14:136-153. [PMID: 32110683 PMCID: PMC7033303 DOI: 10.1016/j.reth.2020.01.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/14/2020] [Accepted: 01/26/2020] [Indexed: 12/14/2022] Open
Abstract
There is enormous global anticipation for stem cell-based therapies that are safe and effective. Numerous pre-clinical studies present encouraging results on the therapeutic potential of different cell types including tissue derived stem cells. Emerging evidences in different fields of research suggest several cell types are safe, whereas their therapeutic application and effectiveness remain challenged. Multiple factors that influence treatment outcomes are proposed including immunocompatibility and potency, owing to variations in tissue origin, ex-vivo methodologies for preparation and handling of the cells. This communication gives an overview of literature data on the different types of cells that are potentially promising for regenerative therapy. As a case in point, the recent trends in research and development of the mesenchymal stem cells (MSCs) for cell therapy are considered in detail. MSCs can be isolated from a variety of tissues and organs in the human body including bone marrow, adipose, synovium, and perinatal tissues. However, MSC products from the different tissue sources exhibit unique or varied levels of regenerative abilities. The review finally focuses on adipose tissue-derived MSCs (ASCs), with the unique properties such as easier accessibility and abundance, excellent proliferation and differentiation capacities, low immunogenicity, immunomodulatory and many other trophic properties. The suitability and application of the ASCs, and strategies to improve the innate regenerative capacities of stem cells in general are highlighted among others.
Collapse
Affiliation(s)
- Edward H. Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Japan
- Research Center for Regenerative Medicine, School of Medicine, University of the Ryukyus, Japan
| | - Hiroshi Sunami
- Research Center for Regenerative Medicine, School of Medicine, University of the Ryukyus, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Japan
| |
Collapse
|
139
|
Huayllani MT, Sarabia-Estrada R, Restrepo DJ, Boczar D, Sisti A, Nguyen JH, Rinker BD, Moran SL, Quiñones-Hinojosa A, Forte AJ. Adipose-derived stem cells in wound healing of full-thickness skin defects: a review of the literature . J Plast Surg Hand Surg 2020; 54:263-279. [PMID: 32427016 DOI: 10.1080/2000656x.2020.1767116] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The complex process of wound healing can be delayed in circumstances when the natural niche is extremely altered. Adipose-derived stem cells (ADSC) seem to be a promising therapy for these type of wounds. We aim to describe the studies that used ADSC for wound healing after a full-thickness skin defect, the ADSC mechanisms of action, and the outcomes of the different ADSC therapies applied to date. We performed a review by querying PubMed database for studies that evaluated the use of ADSC for wound healing. The Mesh terms, adipose stem cells AND (skin injury OR wound healing) and synonyms were used for the search. Our search recorded 312 articles. A total of 30 articles met the inclusion criteria. All were experimental in nature. ADSC was applied directly (5 [16.7%]), in sheets (2 [6.7%]), scaffolds (14 [46.7%]), skin grafts (3 [10%]), skin flaps (1 [3.3%]), as microvesicles or exosomes (4 [13.3%]), with adhesives for wound closure (1 [3.3%]), and in a concentrated conditioned hypoxia-preconditioned medium (1 [3.3%]). Most of the studies reported a benefit of ADSC and improvement of wound healing with all types of ADSC therapy. ADSC applied along with extracellular matrix, stromal cell-derived factor (SDF-1) or keratinocytes, or ADSC seeded in scaffolds showed better outcomes in wound healing than ADSC alone. ADSC have shown to promote angiogenesis, fibroblast migration, and up-regulation of macrophages chemotaxis to enhance the wound healing process. Further studies should be conducted to assure the efficacy and safety of the different ADSC therapies.
Collapse
Affiliation(s)
| | | | | | - Daniel Boczar
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Andrea Sisti
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Justin H Nguyen
- Department of Transplantation Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Brian D Rinker
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Steven L Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
140
|
A Novel 3D Bioprinter Using Direct-Volumetric Drop-On-Demand Technology for Fabricating Micro-Tissues and Drug-Delivery. Int J Mol Sci 2020; 21:ijms21103482. [PMID: 32423161 PMCID: PMC7279004 DOI: 10.3390/ijms21103482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022] Open
Abstract
Drop-on-demand (DOD) 3D bioprinting technologies currently hold the greatest promise for generating functional tissues for clinical use and for drug development. However, existing DOD 3D bioprinting technologies have three main limitations: (1) droplet volume inconsistency; (2) the ability to print only bioinks with low cell concentrations and low viscosity; and (3) problems with cell viability when dispensed under high pressure. We report our success developing a novel direct-volumetric DOD (DVDOD) 3D bioprinting technology that overcomes each of these limitations. DVDOD can produce droplets of bioink from <10 nL in volume using a direct-volumetric mechanism with <± 5% volumetric percent accuracy in an accurate spatially controlled manner. DVDOD has the capability of dispensing bioinks with high concentrations of cells and/or high viscosity biomaterials in either low- or high-throughput modes. The cells are subjected to a low pressure during the bioprinting process for a very short period of time that does not negatively impact cell viability. We demonstrated the functions of the bioprinter in two distinct manners: (1) by using a high-throughput drug-delivery model; and (2) by bioprinting micro-tissues using a variety of different cell types, including functional micro-tissues of bone, cancer, and induced pluripotent stem cells. Our DVDOD technology demonstrates a promising platform for generating many types of tissues and drug-delivery models.
Collapse
|
141
|
Feng L, Liang S, Zhou Y, Luo Y, Chen R, Huang Y, Chen Y, Xu M, Yao R. Three-Dimensional Printing of Hydrogel Scaffolds with Hierarchical Structure for Scalable Stem Cell Culture. ACS Biomater Sci Eng 2020; 6:2995-3004. [DOI: 10.1021/acsbiomaterials.9b01825] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Lu Feng
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Shaojun Liang
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Yongyong Zhou
- Key Laboratory of Medical Information and 3D Bioprinting of Zhejiang Province, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Yixue Luo
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Ruoyu Chen
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Yuyu Huang
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Yiqing Chen
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Mingen Xu
- Key Laboratory of Medical Information and 3D Bioprinting of Zhejiang Province, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Rui Yao
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
142
|
Nicolas J, Magli S, Rabbachin L, Sampaolesi S, Nicotra F, Russo L. 3D Extracellular Matrix Mimics: Fundamental Concepts and Role of Materials Chemistry to Influence Stem Cell Fate. Biomacromolecules 2020; 21:1968-1994. [PMID: 32227919 DOI: 10.1021/acs.biomac.0c00045] [Citation(s) in RCA: 278] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Synthetic 3D extracellular matrices (ECMs) find application in cell studies, regenerative medicine, and drug discovery. While cells cultured in a monolayer may exhibit unnatural behavior and develop very different phenotypes and genotypes than in vivo, great efforts in materials chemistry have been devoted to reproducing in vitro behavior in in vivo cell microenvironments. This requires fine-tuning the biochemical and structural actors in synthetic ECMs. This review will present the fundamentals of the ECM, cover the chemical and structural features of the scaffolds used to generate ECM mimics, discuss the nature of the signaling biomolecules required and exploited to generate bioresponsive cell microenvironments able to induce a specific cell fate, and highlight the synthetic strategies involved in creating functional 3D ECM mimics.
Collapse
Affiliation(s)
- Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, , 92296 Châtenay-Malabry, France
| | - Sofia Magli
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Linda Rabbachin
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Susanna Sampaolesi
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Francesco Nicotra
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Laura Russo
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
143
|
Injectable Therapeutic Organoids Using Sacrificial Hydrogels. iScience 2020; 23:101052. [PMID: 32353766 PMCID: PMC7191221 DOI: 10.1016/j.isci.2020.101052] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/11/2020] [Accepted: 04/03/2020] [Indexed: 12/28/2022] Open
Abstract
Organoids are becoming widespread in drug-screening technologies but have been used sparingly for cell therapy as current approaches for producing self-organized cell clusters lack scalability or reproducibility in size and cellular organization. We introduce a method of using hydrogels as sacrificial scaffolds, which allow cells to form self-organized clusters followed by gentle release, resulting in highly reproducible multicellular structures on a large scale. We demonstrated this strategy for endothelial cells and mesenchymal stem cells to self-organize into blood-vessel units, which were injected into mice, and rapidly formed perfusing vasculature. Moreover, in a mouse model of peripheral artery disease, intramuscular injections of blood-vessel units resulted in rapid restoration of vascular perfusion within seven days. As cell therapy transforms into a new class of therapeutic modality, this simple method—by making use of the dynamic nature of hydrogels—could offer high yields of self-organized multicellular aggregates with reproducible sizes and cellular architectures. Therapeutic, prevascularized organoids were formed in a sacrificial scaffold The organoids are highly reproducible and grown in a high-throughput manner The organoids rapidly formed perfusing vasculature in healthy mice Therapeutic potential was assessed in a mouse model of peripheral artery disease
Collapse
|
144
|
Gargus ES, Rogers HB, McKinnon KE, Edmonds ME, Woodruff TK. Engineered reproductive tissues. Nat Biomed Eng 2020; 4:381-393. [PMID: 32251392 PMCID: PMC7416444 DOI: 10.1038/s41551-020-0525-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 02/05/2020] [Indexed: 12/14/2022]
Abstract
Engineered male and female biomimetic reproductive tissues are being developed as autonomous in vitro units or as integrated multi-organ in vitro systems to support germ cell and embryo function, and to display characteristic endocrine phenotypic patterns, such as the 28-day human ovulatory cycle. In this Review, we summarize how engineered reproductive tissues facilitate research in reproductive biology, and overview strategies for making engineered reproductive tissues that might eventually allow the restoration of reproductive capacity in patients.
Collapse
Affiliation(s)
- Emma S Gargus
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hunter B Rogers
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kelly E McKinnon
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maxwell E Edmonds
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
145
|
Ren T, Steiger W, Chen P, Ovsianikov A, Demirci U. Enhancing cell packing in buckyballs by acoustofluidic activation. Biofabrication 2020; 12:025033. [PMID: 32229710 DOI: 10.1088/1758-5090/ab76d9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
How to pack materials into well-defined volumes efficiently has been a longstanding question of interest to physicists, material scientists, and mathematicians as these materials have broad applications ranging from shipping goods in commerce to seeds in agriculture and to spheroids in tissue engineering. How many marbles or gumball candies can you pack into a jar? Although these seem to be idle questions they have been studied for centuries and have recently become of greater interest with their broadening applications in science and medicine. Here, we study a similar problem where we try to pack cells into a spherical porous buckyball structure. The experimental limitations are short of the theoretical maximum packing density due to the microscale of the structures that the cells are being packed into. We show that we can pack more cells into a confined micro-structure (buckyball cage) by employing acoustofluidic activation and their hydrodynamic effect at the bottom of a liquid-carrier chamber compared to randomly dropping cells onto these buckyballs by gravity. Although, in essence, cells would be expected to achieve a higher maximum volume fraction than marbles in a jar, given that they can squeeze and reshape and reorient their structure, the packing density of cells into the spherical buckyball cages are far from this theoretical limit. This is mainly dictated by the experimental limitations of cells washing away as well as being loaded into the chamber.
Collapse
Affiliation(s)
- Tanchen Ren
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, California 94304, United States of America
| | | | | | | | | |
Collapse
|
146
|
Kronemberger GS, Matsui RAM, Miranda GDASDCE, Granjeiro JM, Baptista LS. Cartilage and bone tissue engineering using adipose stromal/stem cells spheroids as building blocks. World J Stem Cells 2020; 12:110-122. [PMID: 32184936 PMCID: PMC7062040 DOI: 10.4252/wjsc.v12.i2.110] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/19/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Scaffold-free techniques in the developmental tissue engineering area are designed to mimic in vivo embryonic processes with the aim of biofabricating, in vitro, tissues with more authentic properties. Cell clusters called spheroids are the basis for scaffold-free tissue engineering. In this review, we explore the use of spheroids from adult mesenchymal stem/stromal cells as a model in the developmental engineering area in order to mimic the developmental stages of cartilage and bone tissues. Spheroids from adult mesenchymal stromal/stem cells lineages recapitulate crucial events in bone and cartilage formation during embryogenesis, and are capable of spontaneously fusing to other spheroids, making them ideal building blocks for bone and cartilage tissue engineering. Here, we discuss data from ours and other labs on the use of adipose stromal/stem cell spheroids in chondrogenesis and osteogenesis in vitro. Overall, recent studies support the notion that spheroids are ideal "building blocks" for tissue engineering by “bottom-up” approaches, which are based on tissue assembly by advanced techniques such as three-dimensional bioprinting. Further studies on the cellular and molecular mechanisms that orchestrate spheroid fusion are now crucial to support continued development of bottom-up tissue engineering approaches such as three-dimensional bioprinting.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
| | - Renata Akemi Morais Matsui
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
| | - Guilherme de Almeida Santos de Castro e Miranda
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Duque de Caxias, RJ 25250-020, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói 25255-030 Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
147
|
Contessi Negrini N, Lipreri MV, Tanzi MC, Farè S. In vitro cell delivery by gelatin microspheres prepared in water-in-oil emulsion. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:26. [PMID: 32060637 DOI: 10.1007/s10856-020-6363-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 01/27/2020] [Indexed: 06/10/2023]
Abstract
The regeneration of injured or damaged tissues by cell delivery approaches requires the fabrication of cell carriers (e.g., microspheres, MS) that allow for cell delivery to limit cells spreading from the injection site. Ideal MS for cell delivery should allow for cells adhesion and proliferation on the MS before the injection, while they should allow for viable cells release after the injection to promote the damaged tissue regeneration. We optimized a water-in-oil emulsion method to obtain gelatin MS crosslinked by methylenebisacrylamide (MBA). The method we propose allowed obtaining spherical, chemically crosslinked MS characterized by a percentage crosslinking degree of 74.5 ± 2.1%. The chemically crosslinked gelatin MS are characterized by a diameter of 70.9 ± 17.2 μm in the dry state and, at swelling plateau in culture medium at 37 °C, by a diameter of 169.3 ± 41.3 μm. The MS show dimensional stability up to 28 days, after which they undergo complete degradation. Moreover, during their degradation, MS release gelatin that can improve the engraftment of cells in the injured site. The produced MS did not induce any cytotoxic effect in vitro and they supported viable L929 fibroblasts adhesion and proliferation. The MS released viable cells able to colonize and proliferate on the tissue culture plastic, used as release substrate, potentially proving their ability in supporting a simplified in vitro wound healing process, thus representing an optimal tool for cell delivery applications.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy.
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy.
| | - Maria Veronica Lipreri
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Maria Cristina Tanzi
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Silvia Farè
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| |
Collapse
|
148
|
Messina A, Luce E, Hussein M, Dubart-Kupperschmitt A. Pluripotent-Stem-Cell-Derived Hepatic Cells: Hepatocytes and Organoids for Liver Therapy and Regeneration. Cells 2020; 9:cells9020420. [PMID: 32059501 PMCID: PMC7072243 DOI: 10.3390/cells9020420] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/04/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
The liver is a very complex organ that ensures numerous functions; it is thus susceptible to multiple types of damage and dysfunction. Since 1983, orthotopic liver transplantation (OLT) has been considered the only medical solution available to patients when most of their liver function is lost. Unfortunately, the number of patients waiting for OLT is worryingly increasing, and extracorporeal liver support devices are not yet able to counteract the problem. In this review, the current and expected methodologies in liver regeneration are briefly analyzed. In particular, human pluripotent stem cells (hPSCs) as a source of hepatic cells for liver therapy and regeneration are discussed. Principles of hPSC differentiation into hepatocytes are explored, along with the current limitations that have led to the development of 3D culture systems and organoid production. Expected applications of these organoids are discussed with particular attention paid to bio artificial liver (BAL) devices and liver bio-fabrication.
Collapse
Affiliation(s)
- Antonietta Messina
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Eléanor Luce
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Marwa Hussein
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
| | - Anne Dubart-Kupperschmitt
- INSERM unité mixte de recherche (UMR_S) 1193, F-94800 Villejuif, France; (A.M.)
- UMR_S 1193, Université Paris-Sud/Paris-Saclay, F-94800 Villejuif, France
- Département Hospitalo-Universitaire (DHU) Hépatinov, F-94800 Villejuif, France
- Correspondence: ; Tel.: +33-145595138
| |
Collapse
|
149
|
Ivashchenko O, Peplińska B, Przysiecka Ł, Coy E, Jarek M, Chybczyńska K, Jurga S. Nanocomposite Gel as Injectable Therapeutic Scaffold: Microstructural Aspects and Bioactive Properties. ACS APPLIED MATERIALS & INTERFACES 2020; 12:7840-7853. [PMID: 31977186 DOI: 10.1021/acsami.9b23529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The development of tissue scaffolds able to provide proper and accelerated regeneration of tissue is a main task of tissue engineering. We developed a nanocomposite gel that may be used as an injectable therapeutic scaffold. The nanocomposite gel is based on biocompatible gelling agents with embedded nanoparticles (iron oxide, silver, and hydroxyapatite) providing therapeutic properties. We have investigated the microstructure of the nanocomposite gel exposed to different substrates (porous materials and biological tissue). Here we show that the nanocomposite gel has the ability to self-reassemble mimicking the substrate morphology: exposition on porous mineral substrate caused reassembling of nanocomposite gel into 10× smaller scale structure; exposition to a section of humerus cortical bone decreased the microstructure scale more than twice (to ≤3 μm). The reassembling happens through a transitional layer which exists near the phase separation boundary. Our results impact the knowledge of gels explaining their abundance in biological organisms from the microstructural point of view. The results of our biological experiments showed that the nanocomposite gel may find diverse applications in the biomedical field.
Collapse
Affiliation(s)
- Olena Ivashchenko
- NanoBioMedical Centre , Adam Mickiewicz University in Poznań , 61614 Poznań , Poland
| | - Barbara Peplińska
- NanoBioMedical Centre , Adam Mickiewicz University in Poznań , 61614 Poznań , Poland
| | - Łucja Przysiecka
- NanoBioMedical Centre , Adam Mickiewicz University in Poznań , 61614 Poznań , Poland
| | - Emerson Coy
- NanoBioMedical Centre , Adam Mickiewicz University in Poznań , 61614 Poznań , Poland
| | - Marcin Jarek
- NanoBioMedical Centre , Adam Mickiewicz University in Poznań , 61614 Poznań , Poland
| | | | - Stefan Jurga
- NanoBioMedical Centre , Adam Mickiewicz University in Poznań , 61614 Poznań , Poland
| |
Collapse
|
150
|
Zhu L, Luo D, Liu Y. Effect of the nano/microscale structure of biomaterial scaffolds on bone regeneration. Int J Oral Sci 2020; 12:6. [PMID: 32024822 PMCID: PMC7002518 DOI: 10.1038/s41368-020-0073-y] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/15/2019] [Accepted: 12/18/2019] [Indexed: 01/12/2023] Open
Abstract
Natural bone is a mineralized biological material, which serves a supportive and protective framework for the body, stores minerals for metabolism, and produces blood cells nourishing the body. Normally, bone has an innate capacity to heal from damage. However, massive bone defects due to traumatic injury, tumor resection, or congenital diseases pose a great challenge to reconstructive surgery. Scaffold-based tissue engineering (TE) is a promising strategy for bone regenerative medicine, because biomaterial scaffolds show advanced mechanical properties and a good degradation profile, as well as the feasibility of controlled release of growth and differentiation factors or immobilizing them on the material surface. Additionally, the defined structure of biomaterial scaffolds, as a kind of mechanical cue, can influence cell behaviors, modulate local microenvironment and control key features at the molecular and cellular levels. Recently, nano/micro-assisted regenerative medicine becomes a promising application of TE for the reconstruction of bone defects. For this reason, it is necessary for us to have in-depth knowledge of the development of novel nano/micro-based biomaterial scaffolds. Thus, we herein review the hierarchical structure of bone, and the potential application of nano/micro technologies to guide the design of novel biomaterial structures for bone repair and regeneration.
Collapse
Affiliation(s)
- Lisha Zhu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Dan Luo
- State Key Laboratory of Heavy Oil Processing, College of New Energy and Materials, Beijing Key Laboratory of Biogas Upgrading Utilization, China University of Petroleum (Beijing), Beijing, China
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| |
Collapse
|