1
|
Wang W, Liu R, Zhong Q, Cao Y, Qi J, Li Y, Yang Q. Single-cell analysis of nasal epithelial cell development in domestic pigs. Vet Res 2024; 55:140. [PMID: 39478588 PMCID: PMC11523856 DOI: 10.1186/s13567-024-01403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/28/2024] [Indexed: 11/03/2024] Open
Abstract
The nasal mucosa forms a critical barrier against the invasion of respiratory pathogens. Composed of a heterogeneous assortment of cell types, the nasal mucosa relies on the unique characteristics and complex intercellular dynamics of these cells to maintain their structural integrity and functional efficacy. In this study, single-cell RNA sequencing (scRNA-seq) of porcine nasal mucosa was performed, and nineteen distinct nasal cell types, including nine epithelial cell types, five stromal cell types, and five immune cell types, were identified. The distribution patterns of three representative types of epithelial cells (basal cells, goblet cells, and ciliated cells) were subsequently detected by immunofluorescence. We conducted a comparative analysis of these data with published human single-cell data, revealing consistent differentiation trajectories among porcine and human nasal epithelial cells. Specifically, basal cells serve as the initial stage in the differentiation process of nasal epithelial cells, which then epithelial cells. This research not only enhances our understanding of the composition and transcriptional signature of porcine nasal mucosal cells but also offers a theoretical foundation for developing alternative models for human respiratory diseases.
Collapse
Affiliation(s)
- Wenqian Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Ruiling Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Qiu Zhong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yunlei Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jiaxin Qi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China.
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Blatchley MR, Anseth KS. Middle-out methods for spatiotemporal tissue engineering of organoids. NATURE REVIEWS BIOENGINEERING 2023; 1:329-345. [PMID: 37168734 PMCID: PMC10010248 DOI: 10.1038/s44222-023-00039-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/03/2023] [Indexed: 05/13/2023]
Abstract
Organoids recapitulate many aspects of the complex three-dimensional (3D) organization found within native tissues and even display tissue and organ-level functionality. Traditional approaches to organoid culture have largely employed a top-down tissue engineering strategy, whereby cells are encapsulated in a 3D matrix, such as Matrigel, alongside well-defined biochemical cues that direct morphogenesis. However, the lack of spatiotemporal control over niche properties renders cellular processes largely stochastic. Therefore, bottom-up tissue engineering approaches have evolved to address some of these limitations and focus on strategies to assemble tissue building blocks with defined multi-scale spatial organization. However, bottom-up design reduces the capacity for self-organization that underpins organoid morphogenesis. Here, we introduce an emerging framework, which we term middle-out strategies, that relies on existing design principles and combines top-down design of defined synthetic matrices that support proliferation and self-organization with bottom-up modular engineered intervention to limit the degrees of freedom in the dynamic process of organoid morphogenesis. We posit that this strategy will provide key advances to guide the growth of organoids with precise geometries, structures and function, thereby facilitating an unprecedented level of biomimicry to accelerate the utility of organoids to more translationally relevant applications.
Collapse
Affiliation(s)
- Michael R. Blatchley
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| |
Collapse
|
3
|
Tan B, Gan S, Wang X, Liu W, Li X. Applications of 3D bioprinting in tissue engineering: advantages, deficiencies, improvements, and future perspectives. J Mater Chem B 2021; 9:5385-5413. [PMID: 34124724 DOI: 10.1039/d1tb00172h] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the past decade, 3D bioprinting technology has progressed tremendously in the field of tissue engineering in its ability to fabricate individualized biological constructs with precise geometric designability, which offers us the capability to bridge the divergence between engineered tissue constructs and natural tissues. In this work, we first review the current widely used 3D bioprinting approaches, cells, and materials. Next, the updated applications of this technique in tissue engineering, including bone tissue, cartilage tissue, vascular grafts, skin, neural tissue, heart tissue, liver tissue and lung tissue, are briefly introduced. Then, the prominent advantages of 3D bioprinting in tissue engineering are summarized in detail: rapidly prototyping the customized structure, delivering cell-laden materials with high precision in space, and engineering with a highly controllable microenvironment. The current technical deficiencies of 3D bioprinted constructs in terms of mechanical properties and cell behaviors are afterward illustrated, as well as corresponding improvements. Finally, we conclude with future perspectives about 3D bioprinting in tissue engineering.
Collapse
Affiliation(s)
- Baosen Tan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Shaolei Gan
- Jiangxi Borayer Biotech Co., Ltd, Nanchang 330052, China
| | - Xiumei Wang
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Wenyong Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
4
|
Kasendra M, Troutt M, Broda T, Bacon WC, Wang TC, Niland JC, Helmrath MA. Intestinal organoids: roadmap to the clinic. Am J Physiol Gastrointest Liver Physiol 2021; 321:G1-G10. [PMID: 33950707 PMCID: PMC8321798 DOI: 10.1152/ajpgi.00425.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 01/31/2023]
Abstract
Recent advances in intestinal organoid research, along with encouraging preclinical proof-of-concept studies, have revealed significant therapeutic potential for induced pluripotent stem cell (iPSC)-derived organoids in the healing and replacement of severely injured or diseased bowel (Finkbeiner et al. Biol Open 4: 1462-1472, 2015; Kitano et al. Nat Commun 8: 765, 2017; Cruz-Acuna et al. Nat Cell Biol 19: 1326-1335, 2017). To fully realize the tremendous promise of stem cell organoid-based therapies, careful planning aligned with significant resources and efforts must be devoted demonstrating their safety and efficacy to meet critical regulatory requirements. Early recognition of the inherent preclinical and clinical obstacles that occur with the novel use of pluripotent stem cell-derived products will accelerate their bench-to-bedside translation (Neofytou et al. J Clin Invest 125: 2551-2557, 2015; O'Brien et al. Stem Cell Res Ther 6: 146, 2015; Ouseph et al. Cytotherapy 17: 339-343, 2015). To overcome many of these hurdles, a close and effective collaboration is needed between experts from various disciplines, including basic and clinical research, product development and manufacturing, quality assurance and control, and regulatory affairs. Therefore, the purpose of this article is to outline the critical areas and challenges that must be addressed when transitioning laboratory-based discovery, through an investigational new drug (IND) application to first-in-human clinical trial, and to encourage investigators to consider the required regulatory steps from the earliest stage of the translational process. The ultimate goal is to provide readers with a draft roadmap that they could use while navigating this exciting cell therapy space.
Collapse
Affiliation(s)
- Magdalena Kasendra
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Misty Troutt
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Taylor Broda
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - W Clark Bacon
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York City, New York
| | - Joyce C Niland
- Department of Diabetes and Cancer Discovery Science, City of Hope, Duarte, California
| | - Michael A Helmrath
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
5
|
Patino-Guerrero A, Veldhuizen J, Zhu W, Migrino RQ, Nikkhah M. Three-dimensional scaffold-free microtissues engineered for cardiac repair. J Mater Chem B 2020; 8:7571-7590. [PMID: 32724973 PMCID: PMC8314954 DOI: 10.1039/d0tb01528h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases, including myocardial infarction (MI), persist as the leading cause of mortality and morbidity worldwide. The limited regenerative capacity of the myocardium presents significant challenges specifically for the treatment of MI and, subsequently, heart failure (HF). Traditional therapeutic approaches mainly rely on limiting the induced damage or the stress on the remaining viable myocardium through pharmacological regulation of remodeling mechanisms, rather than replacement or regeneration of the injured tissue. The emerging alternative regenerative medicine-based approaches have focused on restoring the damaged myocardial tissue with newly engineered functional and bioinspired tissue units. Cardiac regenerative medicine approaches can be broadly categorized into three groups: cell-based therapies, scaffold-based cardiac tissue engineering, and scaffold-free cardiac tissue engineering. Despite significant advancements, however, the clinical translation of these approaches has been critically hindered by two key obstacles for successful structural and functional replacement of the damaged myocardium, namely: poor engraftment of engineered tissue into the damaged cardiac muscle and weak electromechanical coupling of transplanted cells with the native tissue. To that end, the integration of micro- and nanoscale technologies along with recent advancements in stem cell technologies have opened new avenues for engineering of structurally mature and highly functional scaffold-based (SB-CMTs) and scaffold-free cardiac microtissues (SF-CMTs) with enhanced cellular organization and electromechanical coupling for the treatment of MI and HF. In this review article, we will present the state-of-the-art approaches and recent advancements in the engineering of SF-CMTs for myocardial repair.
Collapse
|
6
|
Kim BS, Das S, Jang J, Cho DW. Decellularized Extracellular Matrix-based Bioinks for Engineering Tissue- and Organ-specific Microenvironments. Chem Rev 2020; 120:10608-10661. [PMID: 32786425 DOI: 10.1021/acs.chemrev.9b00808] [Citation(s) in RCA: 267] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biomaterials-based biofabrication methods have gained much attention in recent years. Among them, 3D cell printing is a pioneering technology to facilitate the recapitulation of unique features of complex human tissues and organs with high process flexibility and versatility. Bioinks, combinations of printable hydrogel and cells, can be utilized to create 3D cell-printed constructs. The bioactive cues of bioinks directly trigger cells to induce tissue morphogenesis. Among the various printable hydrogels, the tissue- and organ-specific decellularized extracellular matrix (dECM) can exert synergistic effects in supporting various cells at any component by facilitating specific physiological properties. In this review, we aim to discuss a new paradigm of dECM-based bioinks able to recapitulate the inherent microenvironmental niche in 3D cell-printed constructs. This review can serve as a toolbox for biomedical engineers who want to understand the beneficial characteristics of the dECM-based bioinks and a basic set of fundamental criteria for printing functional human tissues and organs.
Collapse
Affiliation(s)
- Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu,, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Sanskrita Das
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Jinah Jang
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu,, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
7
|
Wechsler ME, Shevchuk M, Peppas NA. Developing a Multidisciplinary Approach for Engineering Stem Cell Organoids. Ann Biomed Eng 2020; 48:1895-1904. [PMID: 31659603 PMCID: PMC7186139 DOI: 10.1007/s10439-019-02391-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/22/2019] [Indexed: 01/21/2023]
Abstract
Recent advances in stem cell biology, synthetic biology, bioengineering, and biotechnology have included significant work leading to the development of stem cell-derived organoids. The growing popularity of organoid research and use of organoids is widely due to the fact that these three-dimensional cellular structures better model human physiology compared to traditional in vitro and in vivo methods by recapitulating many biologically relevant parameters. Organoids show great promise for a wide range of applications, such as for use in disease modeling, drug discovery, and regenerative medicine. However, many challenges associated with reproducibility and scale up still remain. Identification of the conditions which generate a robust environment that predictably promotes cellular self-assembly and organization leading to organoid formation is critical and requires a multidisciplinary approach. To accomplish this we need to identify a cellular source, engineer a matrix to stimulate cell-cell and cell-matrix interactions, and provide the biochemical and biophysical cues which mimic that of the in vivo environment. Discussion of the components needed for organoid development and formation is reviewed herein, as well as specific organoid examples and the promise of this research for the future.
Collapse
Affiliation(s)
- Marissa E Wechsler
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Mariya Shevchuk
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA.
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.
- Department of Surgery and Perioperative Care, and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
8
|
Gaspar VM, Lavrador P, Borges J, Oliveira MB, Mano JF. Advanced Bottom-Up Engineering of Living Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903975. [PMID: 31823448 DOI: 10.1002/adma.201903975] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/30/2019] [Indexed: 05/08/2023]
Abstract
Bottom-up tissue engineering is a promising approach for designing modular biomimetic structures that aim to recapitulate the intricate hierarchy and biofunctionality of native human tissues. In recent years, this field has seen exciting progress driven by an increasing knowledge of biological systems and their rational deconstruction into key core components. Relevant advances in the bottom-up assembly of unitary living blocks toward the creation of higher order bioarchitectures based on multicellular-rich structures or multicomponent cell-biomaterial synergies are described. An up-to-date critical overview of long-term existing and rapidly emerging technologies for integrative bottom-up tissue engineering is provided, including discussion of their practical challenges and required advances. It is envisioned that a combination of cell-biomaterial constructs with bioadaptable features and biospecific 3D designs will contribute to the development of more robust and functional humanized tissues for therapies and disease models, as well as tools for fundamental biological studies.
Collapse
Affiliation(s)
- Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Pedro Lavrador
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João Borges
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
9
|
Stephenson M, Reich DH, Boheler KR. Induced pluripotent stem cell-derived vascular smooth muscle cells. VASCULAR BIOLOGY 2019; 2:R1-R15. [PMID: 32923972 PMCID: PMC7439844 DOI: 10.1530/vb-19-0028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/12/2019] [Indexed: 12/31/2022]
Abstract
The reproducible generation of human-induced pluripotent stem cell (hiPSC)-derived vascular smooth muscle cells (vSMCs) in vitro has been critical to overcoming many limitations of animal and primary cell models of vascular biology and disease. Since this initial advance, research in the field has turned toward recapitulating the naturally occurring subtype specificity found in vSMCs throughout the body, and honing functional models of vascular disease. In this review, we summarize vSMC derivation approaches, including current phenotype and developmental origin-specific methods, and applications of vSMCs in functional disease models and engineered tissues. Further, we discuss the challenges of heterogeneity in hiPSC-derived tissues and propose approaches to identify and isolate vSMC subtype populations.
Collapse
Affiliation(s)
- Makeda Stephenson
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel H Reich
- Department of Physics and Astronomy, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Kenneth R Boheler
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Xu Y, Chen C, Hellwarth PB, Bao X. Biomaterials for stem cell engineering and biomanufacturing. Bioact Mater 2019; 4:366-379. [PMID: 31872161 PMCID: PMC6909203 DOI: 10.1016/j.bioactmat.2019.11.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Recent years have witnessed the expansion of tissue failures and diseases. The uprising of regenerative medicine converges the sight onto stem cell-biomaterial based therapy. Tissue engineering and regenerative medicine proposes the strategy of constructing spatially, mechanically, chemically and biologically designed biomaterials for stem cells to grow and differentiate. Therefore, this paper summarized the basic properties of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. The properties of frequently used biomaterials were also described in terms of natural and synthetic origins. Particularly, the combination of stem cells and biomaterials for tissue repair applications was reviewed in terms of nervous, cardiovascular, pancreatic, hematopoietic and musculoskeletal system. Finally, stem-cell-related biomanufacturing was envisioned and the novel biofabrication technologies were discussed, enlightening a promising route for the future advancement of large-scale stem cell-biomaterial based therapeutic manufacturing.
Collapse
Affiliation(s)
| | | | | | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, West Lafayette, IN, 47907, USA
| |
Collapse
|
11
|
Libby AR, Joy DA, So PL, Mandegar MA, Muncie JM, Mendoza-Camacho FN, Weaver VM, Conklin BR, McDevitt TC. Spatiotemporal mosaic self-patterning of pluripotent stem cells using CRISPR interference. eLife 2018; 7:36045. [PMID: 30298816 PMCID: PMC6177255 DOI: 10.7554/elife.36045] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 09/09/2018] [Indexed: 12/22/2022] Open
Abstract
Morphogenesis involves interactions of asymmetric cell populations to form complex multicellular patterns and structures comprised of distinct cell types. However, current methods to model morphogenic events lack control over cell-type co-emergence and offer little capability to selectively perturb specific cell subpopulations. Our in vitro system interrogates cell-cell interactions and multicellular organization within human induced pluripotent stem cell (hiPSC) colonies. We examined effects of induced mosaic knockdown of molecular regulators of cortical tension (ROCK1) and cell-cell adhesion (CDH1) with CRISPR interference. Mosaic knockdown of ROCK1 or CDH1 resulted in differential patterning within hiPSC colonies due to cellular self-organization, while retaining an epithelial pluripotent phenotype. Knockdown induction stimulates a transient wave of differential gene expression within the mixed populations that stabilized in coordination with observed self-organization. Mosaic patterning enables genetic interrogation of emergent multicellular properties, which can facilitate better understanding of the molecular pathways that regulate symmetry-breaking during morphogenesis. Embryos begin as a collection of similar cells, which progress in stages to form a huge variety of cell types in particular arrangements. These patterns of cells give rise to the different tissues and organs that make up the body. Although we often use ‘model’ organisms such as mice and frogs to study how embryos develop, our species has evolved unique ways to control organ development. Investigating these processes is difficult: we cannot experiment on human embryos, and our development is hard to recreate in test tubes. As a result, we do not fully understand how developing human cells specialize and organize. Libby et al. have now created a new system to study how different genes control cell organization. The system uses human pluripotent stem cells – cells that have the ability to specialize into any type of cell. Some of the stem cells are modified using a technique called inducible CRISPR interference, which makes it possible to reduce the activity of certain genes in these cells. Libby et al. used this technique to investigate how changes to the activity of two genes – called ROCK1 and CDH1 – affect how a mixed group of stem cells organized themselves. Cells that lacked ROCK1 formed bands near the edges of the group. Cells that lacked CDH1 segregated themselves from other cells, forming ‘islands’ inside the main group. The cells retained their ability to specialize into any type of cell after forming these patterns. However, specific groups of cells were more likely to become certain cell types. The method developed by Libby et al. can be used to study a range of complex tissue development and cell organization processes. Future work could create human tissue model systems for research into human disease or drug development.
Collapse
Affiliation(s)
- Ashley Rg Libby
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, United States
| | - David A Joy
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Graduate Program in Bioengineering, University of California Berkeley, University of California San Francisco, San Francisco, United States
| | - Po-Lin So
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States
| | | | - Jonathon M Muncie
- Graduate Program in Bioengineering, University of California Berkeley, University of California San Francisco, San Francisco, United States.,Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, United States
| | | | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, United States
| | - Bruce R Conklin
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Department of Medicine, Division of Genomic Medicine, University of California, San Francisco, United States
| | - Todd C McDevitt
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, United States
| |
Collapse
|
12
|
Joshi R, Thakuri PS, Buchanan JC, Li J, Tavana H. Microprinted Stem Cell Niches Reveal Compounding Effect of Colony Size on Stromal Cells-Mediated Neural Differentiation. Adv Healthc Mater 2018; 7:10.1002/adhm.201700832. [PMID: 29193846 PMCID: PMC5842135 DOI: 10.1002/adhm.201700832] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/02/2017] [Indexed: 01/30/2023]
Abstract
Microenvironmental factors have a major impact on differentiation of embryonic stem cells (ESCs). Here, a novel phenomenon that size of ESC colonies has a significant regulatory role on stromal cells induced differentiation of ESCs to neural cells is reported. Using a robotic cell microprinting technology, defined densities of ESCs are confined within aqueous nanodrops over a layer of supporting stromal cells immersed in a second, immiscible aqueous phase to generate ESC colonies of defined sizes. Temporal protein and gene expression studies demonstrate that larger ESC colonies generate disproportionally more neural cells and longer neurite processes. Unlike previous studies that attribute neural differentiation of ESCs solely to interactions with stromal cells, it is found that increased intercellular signaling of ESCs significantly enhances neural differentiation. This study offers an approach to generate neural cells with improved efficiency for potential use in translational research.
Collapse
Affiliation(s)
- Ramila Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - James C Buchanan
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Jun Li
- Department of Mathematical Sciences, Kent State University, Kent, OH, 44242, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, 260 S. Forge St., Akron, OH, 44325, USA
| |
Collapse
|
13
|
Laurent J, Blin G, Chatelain F, Vanneaux V, Fuchs A, Larghero J, Théry M. Convergence of microengineering and cellular self-organization towards functional tissue manufacturing. Nat Biomed Eng 2017; 1:939-956. [DOI: 10.1038/s41551-017-0166-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
|
14
|
Johnson MB, March AR, Morsut L. Engineering multicellular systems: using synthetic biology to control tissue self-organization. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 4:163-173. [PMID: 29308442 DOI: 10.1016/j.cobme.2017.10.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Marion B Johnson
- The Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine University of Southern California Keck School of Medicine 1425 San Pablo Avenue, BCC-507, Los Angeles, 90033, USA
| | - Alexander R March
- The Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine University of Southern California Keck School of Medicine 1425 San Pablo Avenue, BCC-507, Los Angeles, 90033, USA
| | - Leonardo Morsut
- The Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine University of Southern California Keck School of Medicine 1425 San Pablo Avenue, BCC-507, Los Angeles, 90033, USA
| |
Collapse
|
15
|
Du V, Luciani N, Richard S, Mary G, Gay C, Mazuel F, Reffay M, Menasché P, Agbulut O, Wilhelm C. A 3D magnetic tissue stretcher for remote mechanical control of embryonic stem cell differentiation. Nat Commun 2017; 8:400. [PMID: 28900152 PMCID: PMC5596024 DOI: 10.1038/s41467-017-00543-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
The ability to create a 3D tissue structure from individual cells and then to stimulate it at will is a major goal for both the biophysics and regenerative medicine communities. Here we show an integrated set of magnetic techniques that meet this challenge using embryonic stem cells (ESCs). We assessed the impact of magnetic nanoparticles internalization on ESCs viability, proliferation, pluripotency and differentiation profiles. We developed magnetic attractors capable of aggregating the cells remotely into a 3D embryoid body. This magnetic approach to embryoid body formation has no discernible impact on ESC differentiation pathways, as compared to the hanging drop method. It is also the base of the final magnetic device, composed of opposing magnetic attractors in order to form embryoid bodies in situ, then stretch them, and mechanically stimulate them at will. These stretched and cyclic purely mechanical stimulations were sufficient to drive ESCs differentiation towards the mesodermal cardiac pathway. The development of embryoid bodies that are responsive to external stimuli is of great interest in tissue engineering. Here, the authors culture embryonic stem cells with magnetic nanoparticles and show that the presence of magnetic fields could affect their aggregation and differentiation.
Collapse
Affiliation(s)
- Vicard Du
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Nathalie Luciani
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Sophie Richard
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Gaëtan Mary
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Cyprien Gay
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - François Mazuel
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Myriam Reffay
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou; Paris Cardiovascular Research Center, INSERM U970, Université Paris Descartes, Paris, 75015, France
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, 75005, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France.
| |
Collapse
|
16
|
Arora N, Imran Alsous J, Guggenheim JW, Mak M, Munera J, Wells JM, Kamm RD, Asada HH, Shvartsman SY, Griffith LG. A process engineering approach to increase organoid yield. Development 2017; 144:1128-1136. [PMID: 28174251 DOI: 10.1242/dev.142919] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/25/2017] [Indexed: 12/15/2022]
Abstract
Temporal manipulation of the in vitro environment and growth factors can direct differentiation of human pluripotent stem cells into organoids - aggregates with multiple tissue-specific cell types and three-dimensional structure mimicking native organs. A mechanistic understanding of early organoid formation is essential for improving the robustness of these methods, which is necessary prior to use in drug development and regenerative medicine. We investigated intestinal organoid emergence, focusing on measurable parameters of hindgut spheroids, the intermediate step between definitive endoderm and mature organoids. We found that 13% of spheroids were pre-organoids that matured into intestinal organoids. Spheroids varied by several structural parameters: cell number, diameter and morphology. Hypothesizing that diameter and the morphological feature of an inner mass were key parameters for spheroid maturation, we sorted spheroids using an automated micropipette aspiration and release system and monitored the cultures for organoid formation. We discovered that populations of spheroids with a diameter greater than 75 μm and an inner mass are enriched 1.5- and 3.8-fold for pre-organoids, respectively, thus providing rational guidelines towards establishing a robust protocol for high quality intestinal organoids.
Collapse
Affiliation(s)
- Natasha Arora
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Jasmin Imran Alsous
- Department of Chemical and Biological Engineering, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Jacob W Guggenheim
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Michael Mak
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Jorge Munera
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA.,Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - H Harry Asada
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Stanislav Y Shvartsman
- Department of Chemical and Biological Engineering, Princeton University, Washington Road, Princeton, NJ 08540, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA .,Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| |
Collapse
|