1
|
Nosrati H, Fallah Tafti M, Aghamollaei H, Bonakdar S, Moosazadeh Moghaddam M. Directed Differentiation of Adipose-Derived Stem Cells Using Imprinted Cell-Like Topographies as a Growth Factor-Free Approach. Stem Cell Rev Rep 2024; 20:1752-1781. [PMID: 39066936 DOI: 10.1007/s12015-024-10767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
The influence of surface topography on stem cell behavior and differentiation has garnered significant attention in regenerative medicine and tissue engineering. The cell-imprinting method has been introduced as a promising approach to mimic the geometry and topography of cells. The cell-imprinted substrates are designed to replicate the topographies and dimensions of target cells, enabling tailored interactions that promote the differentiation of stem cells towards desired specialized cell types. In fact, by replicating the size and shape of cells, biomimetic substrates provide physical cues that profoundly impact stem cell differentiation. These cues play a pivotal role in directing cell morphology, cytoskeletal organization, and gene expression, ultimately influencing lineage commitment. The biomimetic substrates' ability to emulate the native cellular microenvironment supports the creation of platforms capable of steering stem cell fate with high precision. This review discusses the role of mechanical factors that impact stem cell fate. It also provides an overview of the design and fabrication principles of cell-imprinted substrates. Furthermore, the paper delves into the use of cell-imprinted polydimethylsiloxane (PDMS) substrates to direct adipose-derived stem cells (ADSCs) differentiation into a variety of specialized cells for tissue engineering and regenerative medicine applications. Additionally, the review discusses the limitations of cell-imprinted PDMS substrates and highlights the efforts made to overcome these limitations.
Collapse
Affiliation(s)
- Hamed Nosrati
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahsa Fallah Tafti
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran.
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Lin Q, Yang Z, Xu H, Niu Y, Meng Q, Xing D. Advances in Shear Stress Stimulation of Stem Cells: A Review of the Last Three Decades. Biomedicines 2024; 12:1963. [PMID: 39335477 PMCID: PMC11429308 DOI: 10.3390/biomedicines12091963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/30/2024] Open
Abstract
Stem cells are widely used in scientific research because of their ability to self-renew and differentiate into a variety of specialized cell types needed for body functions. However, the self-renewal and differentiation of stem cells are regulated by various stimuli, with mechanical stimulation being particularly notable due to its ability to mimic the physical environment in the body. This study systematically collected 2638 research papers published between 1994 and 2024, employing tools such as VOSviewer, CiteSpace, and GraphPad Prism to uncover research hotspots, publication trends, and collaboration networks. The results indicate a yearly increase in global research on the shear stress stimulation of stem cells, with significant contributions from the United States and China in terms of research investment and output. Future research directions include a deeper understanding of the mechanisms underlying mechanical stimulation's effects on stem cell differentiation, the development of new materials and scaffold designs to better replicate the natural cellular environment, and advancements in regenerative medicine. Despite considerable progress, challenges remain in translating basic research findings into clinical applications.
Collapse
Affiliation(s)
- Qiyuan Lin
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Hao Xu
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Yudi Niu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qingchen Meng
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Dan Xing
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| |
Collapse
|
3
|
Borys BS, Dang T, Worden H, Larijani L, Corpuz JM, Abraham BD, Gysel EJ, Malinovska J, Krawetz R, Revay T, Argiropoulos B, Rancourt DE, Kallos MS, Jung S. Robust bioprocess design and evaluation of commercial media for the serial expansion of human induced pluripotent stem cell aggregate cultures in vertical-wheel bioreactors. Stem Cell Res Ther 2024; 15:232. [PMID: 39075528 PMCID: PMC11288049 DOI: 10.1186/s13287-024-03819-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND While pluripotent stem cell (PSC) therapies move toward clinical and commercial applications at a rapid rate, manufacturing reproducibility and robustness are notable bottlenecks in regulatory approval. Therapeutic applications of PSCs require large cell quantities to be generated under highly robust, well-defined, and economically viable conditions. Small-scale and short-term process optimization, however, is often performed in a linear fashion that does not account for time needed to verify the bioprocess protocols and analysis methods used. Design of a reproducible and robust bioprocess should be dynamic and include a continuous effort to understand how the process will respond over time and to different stresses before transitioning into large-scale production where stresses will be amplified. METHODS This study utilizes a baseline protocol, developed for the short-term culture of PSC aggregates in Vertical-Wheel® bioreactors, to evaluate key process attributes through long-term (serial passage) suspension culture. This was done to access overall process robustness when performed with various commercially available media and cell lines. Process output variables including growth kinetics, aggregate morphology, harvest efficiency, genomic stability, and functional pluripotency were assessed through short and long-term culture. RESULTS The robust nature of the expansion protocol was demonstrated over a six-day culture period where spherical aggregate formation and expansion were observed with high-fold expansions for all five commercial media tested. Profound differences in cell growth and quality were revealed only through long-term serial expansion and in-vessel dissociation operations. Some commercial media formulations tested demonstrated maintenance of cell growth rates, aggregate morphology, and high harvest recovery efficiencies through three bioreactor serial passages using multiple PSC lines. Exceptional bioprocess robustness was even demonstrated with sustained growth and quality maintenance over 10 serial bioreactor passages. However, some commercial media tested proved less equipped for serial passage cultures in bioreactors as cultures led to cell lysis during dissociation, reduction in growth rates, and a loss of aggregate morphology. CONCLUSIONS This study demonstrates the importance of systematic selection and testing of bioprocess input variables, with multiple bioprocess output variables through serial passages to create a truly reproducible and robust protocol for clinical and commercial PSC production using scalable bioreactor systems.
Collapse
Affiliation(s)
- Breanna S Borys
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- PBS Biotech Inc, 4721 Calle Carga, Camarillo, CA, 93012, USA
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Hannah Worden
- PBS Biotech Inc, 4721 Calle Carga, Camarillo, CA, 93012, USA
| | - Leila Larijani
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Jessica M Corpuz
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Brett D Abraham
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Emilie J Gysel
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Julia Malinovska
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Roman Krawetz
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Tamas Revay
- Department of Medical Genetics, Alberta Health Services, Alberta Children's Hospital, Calgary, AB, Canada
| | - Bob Argiropoulos
- Department of Medical Genetics, Alberta Health Services, Alberta Children's Hospital, Calgary, AB, Canada
| | - Derrick E Rancourt
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Sunghoon Jung
- PBS Biotech Inc, 4721 Calle Carga, Camarillo, CA, 93012, USA.
| |
Collapse
|
4
|
Wang N, Chen J, Hu Q, He Y, Shen P, Yang D, Wang H, Weng D, He Z. Small diameter vascular grafts: progress on electrospinning matrix/stem cell blending approach. Front Bioeng Biotechnol 2024; 12:1385032. [PMID: 38807647 PMCID: PMC11130446 DOI: 10.3389/fbioe.2024.1385032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024] Open
Abstract
The exploration of the next-generation small diameter vascular grafts (SDVGs) will never stop until they possess high biocompatibility and patency comparable to autologous native blood vessels. Integrating biocompatible electrospinning (ES) matrices with highly bioactive stem cells (SCs) provides a rational and promising solution. ES is a simple, fast, flexible and universal technology to prepare extracellular matrix-like fibrous scaffolds in large scale, while SCs are valuable, multifunctional and favorable seed cells with special characteristics for the emerging field of cell therapy and regenerative medicine. Both ES matrices and SCs are advanced resources with medical application prospects, and the combination may share their advantages to drive the overcoming of the long-lasting hurdles in SDVG field. In this review, the advances on SDVGs based on ES matrices and SCs (including pluripotent SCs, multipotent SCs, and unipotent SCs) are sorted out, and current challenges and future prospects are discussed.
Collapse
Affiliation(s)
- Nuoxin Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Jiajing Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Qingqing Hu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Yunfeng He
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Pu Shen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Dingkun Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Haoyuan Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Second Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Dong Weng
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Zhixu He
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
5
|
Komosa ER, Lin WH, Mahadik B, Bazzi MS, Townsend D, Fisher JP, Ogle BM. A novel perfusion bioreactor promotes the expansion of pluripotent stem cells in a 3D-bioprinted tissue chamber. Biofabrication 2023; 16:014101. [PMID: 37906964 PMCID: PMC10636629 DOI: 10.1088/1758-5090/ad084a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/02/2023]
Abstract
While the field of tissue engineering has progressed rapidly with the advent of 3D bioprinting and human induced pluripotent stem cells (hiPSCs), impact is limited by a lack of functional, thick tissues. One way around this limitation is to 3D bioprint tissues laden with hiPSCs. In this way, the iPSCs can proliferate to populate the thick tissue mass prior to parenchymal cell specification. Here we design a perfusion bioreactor for an hiPSC-laden, 3D-bioprinted chamber with the goal of proliferating the hiPSCs throughout the structure prior to differentiation to generate a thick tissue model. The bioreactor, fabricated with digital light projection, was optimized to perfuse the interior of the hydrogel chamber without leaks and to provide fluid flow around the exterior as well, maximizing nutrient delivery throughout the chamber wall. After 7 days of culture, we found that intermittent perfusion (15 s every 15 min) at 3 ml min-1provides a 1.9-fold increase in the density of stem cell colonies in the engineered tissue relative to analogous chambers cultured under static conditions. We also observed a more uniform distribution of colonies within the tissue wall of perfused structures relative to static controls, reflecting a homogeneous distribution of nutrients from the culture media. hiPSCs remained pluripotent and proliferative with application of fluid flow, which generated wall shear stresses averaging ∼1.0 dyn cm-2. Overall, these promising outcomes following perfusion of a stem cell-laden hydrogel support the production of multiple tissue types with improved thickness, and therefore increased function and utility.
Collapse
Affiliation(s)
- Elizabeth R Komosa
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States of America
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
| | - Wei-Han Lin
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Bhushan Mahadik
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
- Fishell Department of Bioengineering, University of Maryland, College Park, MD, United States of America
| | - Marisa S Bazzi
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, United States of America
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States of America
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - John P Fisher
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
- Fishell Department of Bioengineering, University of Maryland, College Park, MD, United States of America
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States of America
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States of America
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
6
|
Bakhshandeh B, Sorboni SG, Ranjbar N, Deyhimfar R, Abtahi MS, Izady M, Kazemi N, Noori A, Pennisi CP. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp Cell Res 2023; 431:113766. [PMID: 37678504 DOI: 10.1016/j.yexcr.2023.113766] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Roham Deyhimfar
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Sadat Abtahi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrnaz Izady
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Navid Kazemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Atefeh Noori
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
7
|
Born G, Plantier E, Nannini G, Caimi A, Mazzoleni A, Asnaghi MA, Muraro MG, Scherberich A, Martin I, García-García A. Mini- and macro-scale direct perfusion bioreactors with optimized flow for engineering 3D tissues. Biotechnol J 2023; 18:e2200405. [PMID: 36428229 DOI: 10.1002/biot.202200405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/17/2022] [Accepted: 10/27/2022] [Indexed: 11/28/2022]
Abstract
Bioreactors enabling direct perfusion of cell suspensions or culture media through the pores of 3D scaffolds have long been used in tissue engineering to improve cell seeding efficiency as well as uniformity of cell distribution and tissue development. A macro-scale U-shaped bioreactor for cell culture under perfusion (U-CUP) has been previously developed. In that system, the geometry of the perfusion chamber results in rather uniform flow through most of the scaffold volume, but not in the peripheral regions. Here, the design of the perfusion chamber has been optimized to provide a more homogenous perfusion flow through the scaffold. Then, the design of this macro-scale flow-optimized perfusion bioreactor (macro-Flopper) has been miniaturized to create a mini-scale device (mini-Flopper) compatible with medium-throughput assays. Computational fluid dynamic (CFD) modeling of the new chamber design, including a porous scaffold structure, revealed that Flopper bioreactors provide highly homogenous flow speed, pressure, and shear stress. Finally, a proof-of-principle of the functionality of the Flopper systems by engineering endothelialized stromal tissues using human adipose tissue-derived stromal vascular fraction (SVF) cells has been offered. Preliminary evidence showing that flow optimization improves cell maintenance in the engineered tissues will have to be confirmed in future studies. In summary, two bioreactor models with optimized perfusion flow and complementary sizes have been proposed that might be exploited to engineer homogenous tissues and, in the case of the mini-Flopper, for drug testing assays with a limited amount of biological material.
Collapse
Affiliation(s)
- Gordian Born
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Evelia Plantier
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Guido Nannini
- Department of Electronics, Informatics and Bioengineering (DEIB), Politecnico di Milano, Milan, MI, Italy
| | - Alessandro Caimi
- Department of Electronics, Informatics and Bioengineering (DEIB), Politecnico di Milano, Milan, MI, Italy
| | - Andrea Mazzoleni
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - M Adelaide Asnaghi
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Manuele G Muraro
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrés García-García
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
8
|
Riquelme-Guzmán C, Beck T, Edwards-Jorquera S, Schlüßler R, Müller P, Guck J, Möllmert S, Sandoval-Guzmán T. In vivo assessment of mechanical properties during axolotl development and regeneration using confocal Brillouin microscopy. Open Biol 2022; 12:220078. [PMID: 35728623 PMCID: PMC9213112 DOI: 10.1098/rsob.220078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In processes such as development and regeneration, where large cellular and tissue rearrangements occur, cell fate and behaviour are strongly influenced by tissue mechanics. While most well-established tools probing mechanical properties require an invasive sample preparation, confocal Brillouin microscopy captures mechanical parameters optically with high resolution in a contact-free and label-free fashion. In this work, we took advantage of this tool and the transparency of the highly regenerative axolotl to probe its mechanical properties in vivo for the first time. We mapped the Brillouin frequency shift with high resolution in developing limbs and regenerating digits, the most studied structures in the axolotl. We detected a gradual increase in the cartilage Brillouin frequency shift, suggesting decreasing tissue compressibility during both development and regeneration. Moreover, we were able to correlate such an increase with the regeneration stage, which was undetected with fluorescence microscopy imaging. The present work evidences the potential of Brillouin microscopy to unravel the mechanical changes occurring in vivo in axolotls, setting the basis to apply this technique in the growing field of epimorphic regeneration.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- CRTD/Center for Regenerative Therapies TU Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Timon Beck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sandra Edwards-Jorquera
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Raimund Schlüßler
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Paul Müller
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Stephanie Möllmert
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Tatiana Sandoval-Guzmán
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Paul Langerhans Institute Dresden, Helmholtz Centre Munich, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Kuang J, Sun W, Zhang M, Kang L, Yang S, Zhang H, Wang Y, Hu P. A three-dimensional biomimetic microfluidic chip to study the behavior of hepatic stellate cell under the tumor microenvironment. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
10
|
Liu B, Wang X, Jiang L, Xu J, Zohar Y, Yao G. Extracellular Fluid Flow Induces Shallow Quiescence Through Physical and Biochemical Cues. Front Cell Dev Biol 2022; 10:792719. [PMID: 35281101 PMCID: PMC8912726 DOI: 10.3389/fcell.2022.792719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The balance between cell quiescence and proliferation is fundamental to tissue physiology and homeostasis. Recent studies have shown that quiescence is not a passive and homogeneous state but actively maintained and heterogeneous. These cellular characteristics associated with quiescence were observed primarily in cultured cells under a static medium. However, cells in vivo face different microenvironmental conditions, particularly, under interstitial fluid flows distributed through extracellular matrices. Interstitial fluid flow exerts shear stress on cells and matrix strain, and results in continuous replacement of extracellular factors. In this study, we analyzed individual cells under varying fluid flow rates in microfluidic devices. We found quiescence characteristics previously identified under conventional static medium, including serum signal-dependant quiescence entry and exit and time-dependant quiescence deepening, are also present under continuous fluid flow. Furthermore, increasing the flow rate drives cells to shallower quiescence and become more likely to reenter the cell cycle upon growth stimulation. This effect is due to flow-induced physical and biochemical cues. Specifically, increasing shear stress or extracellular factor replacement individually, without altering other parameters, results in shallow quiescence. We show our experimental results can be quantitatively explained by a mathematical model connecting extracellular fluid flow to an Rb-E2f bistable switch that regulates the quiescence-to-proliferation transition. Our findings uncover a previously unappreciated mechanism that likely underlies the heterogeneous responses of quiescent cells for tissue repair and regeneration in different physiological tissue microenvironments.
Collapse
Affiliation(s)
- Bi Liu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, China
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Xia Wang
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Linan Jiang
- Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, United States
- *Correspondence: Linan Jiang, ; Guang Yao,
| | - Jianhua Xu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, China
| | - Yitshak Zohar
- Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, United States
| | - Guang Yao
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
- *Correspondence: Linan Jiang, ; Guang Yao,
| |
Collapse
|
11
|
Rivera-Ordaz A, Peli V, Manzini P, Barilani M, Lazzari L. Critical Analysis of cGMP Large-Scale Expansion Process in Bioreactors of Human Induced Pluripotent Stem Cells in the Framework of Quality by Design. BioDrugs 2021; 35:693-714. [PMID: 34727354 PMCID: PMC8561684 DOI: 10.1007/s40259-021-00503-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 10/28/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are manufactured as advanced therapy medicinal products for tissue replacement applications. With this aim, the feasibility of hiPSC large-scale expansion in existing bioreactor systems under current good manufacturing practices (cGMP) has been tested. Yet, these attempts have lacked a paradigm shift in culture settings and technologies tailored to hiPSCs, which jeopardizes their clinical translation. The best approach for industrial scale-up of high-quality hiPSCs is to design their manufacturing process by following quality-by-design (QbD) principles: a scientific, risk-based framework for process design based on relating product and process attributes to product quality. In this review, we analyzed the hiPSC expansion manufacturing process implementing the QbD approach in the use of bioreactors, stressing the decisive role played by the cell quantity, quality and costs, drawing key QbD concepts directly from the guidelines of the International Council for Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use.
Collapse
Affiliation(s)
- Araceli Rivera-Ordaz
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Valeria Peli
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Paolo Manzini
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Mario Barilani
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| |
Collapse
|
12
|
Huang Y, Qian JY, Cheng H, Li XM. Effects of shear stress on differentiation of stem cells into endothelial cells. World J Stem Cells 2021; 13:894-913. [PMID: 34367483 PMCID: PMC8316872 DOI: 10.4252/wjsc.v13.i7.894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell transplantation is an appealing potential therapy for vascular diseases and an indispensable key step in vascular tissue engineering. Substantial effort has been made to differentiate stem cells toward vascular cell phenotypes, including endothelial cells (ECs) and smooth muscle cells. The microenvironment of vascular cells not only contains biochemical factors that influence differentiation but also exerts hemodynamic forces, such as shear stress and cyclic strain. More recently, studies have shown that shear stress can influence the differentiation of stem cells toward ECs. A deep understanding of the responses and underlying mechanisms involved in this process is essential for clinical translation. This review highlights current data supporting the role of shear stress in stem cell differentiation into ECs. Potential mechanisms and signaling cascades for transducing shear stress into a biological signal are proposed. Further study of stem cell responses to shear stress will be necessary to apply stem cells for pharmacological applications and cardiovascular implants in the realm of regenerative medicine.
Collapse
Affiliation(s)
- Yan Huang
- 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
| | - Jia-Yi Qian
- 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
| | - Hong Cheng
- 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
| | - Xiao-Ming 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
|
13
|
Hayward MK, Muncie JM, Weaver VM. Tissue mechanics in stem cell fate, development, and cancer. Dev Cell 2021; 56:1833-1847. [PMID: 34107299 PMCID: PMC9056158 DOI: 10.1016/j.devcel.2021.05.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022]
Abstract
Cells in tissues experience a plethora of forces that regulate their fate and modulate development and homeostasis. Cells sense mechanical cues through localized mechanoreceptors or by influencing cytoskeletal or plasma membrane organization. Cells translate force and modulate their behavior through a process termed mechanotransduction. Cells tune their tension upon exposure to chronic force by engaging cellular machinery that modulates actin tension, which in turn stimulates matrix remodeling and stiffening and alters cell-cell adhesions until cells achieve a state of tensional homeostasis. Loss of tensional homeostasis can be induced through oncogene activity and/or tissue fibrosis, accompanies tumor progression, and is associated with increased cancer risk. The mechanical stresses that develop in tumors can also foster the mesenchymal-like transdifferentiation of cells to induce a stem-like phenotype that contributes to their aggression, metastatic dissemination, and treatment resistance. Thus, strategies that ameliorate tumor mechanics may comprise an effective strategy to prevent aggressive tumor behavior.
Collapse
Affiliation(s)
- Mary-Kate Hayward
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences and Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
14
|
Li Z, Li JN, Li Q, Liu C, Zhou LH, Zhang Q, Xu Y. miR-25-5p regulates endothelial progenitor cell differentiation in response to shear stress through targeting ABCA1. Cell Biol Int 2021; 45:1876-1886. [PMID: 33945659 DOI: 10.1002/cbin.11621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/07/2021] [Accepted: 05/01/2021] [Indexed: 11/10/2022]
Abstract
The importance of flow shear stress (SS) on the differentiation of endothelial progenitor cells (EPCs) has been demonstrated in various studies. Cholesterol retention and microRNA regulation have been also proposed as relevant factors involved in this process, though evidence regarding their regulatory roles in the differentiation of EPCs is currently lacking. In the present study on high shear stress (HSS)-induced differentiation of EPCs, we investigated the importance of ATP-binding cassette transporter 1 (ABCA1), an important regulator in cholesterol efflux, and miR-25-5p, a potential regulator of endothelial reconstruction. We first revealed an inverse correlation between miR-25-5p and ABCA1 expression levels in EPCs under HSS treatment; their direct interaction was subsequently validated by a dual-luciferase reporter assay. Further studies using flow cytometry and quantitative polymerase chain reaction demonstrated that both miR-25-5p overexpression and ABCA1 inhibition led to elevated levels of specific markers of endothelial cells, with concomitant downregulation of smooth muscle cell markers. Finally, knockdown of ABCA1 in EPCs significantly promoted tube formation, which confirmed our conjecture. Our current results suggest that miR-25-5p might regulate the differentiation of EPCs partially through targeting ABCA1, and such a mechanism might account for HSS-induced differentiation of EPCs.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Jia-Nan Li
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Qiang Li
- Department of Neurosurgery, Changhai Hospital of Shanghai affiliated to Naval Military Medical University, Shanghai, China
| | - Chun Liu
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Lin-Hua Zhou
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Qi Zhang
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital affiliated to Tongji University, Shanghai, China
| | - Yi Xu
- Department of Neurosurgery, Changhai Hospital of Shanghai affiliated to Naval Military Medical University, Shanghai, China
| |
Collapse
|
15
|
Response of Pluripotent Stem Cells to Environmental Stress and Its Application for Directed Differentiation. BIOLOGY 2021; 10:biology10020084. [PMID: 33498611 PMCID: PMC7912122 DOI: 10.3390/biology10020084] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Environmental changes in oxygen concentration, temperature, and mechanical stimulation lead to the activation of specific transcriptional factors and induce the expression of each downstream gene. In general, these responses are protective machinery against such environmental stresses, while these transcriptional factors also regulate cell proliferation, differentiation, and organ development in mammals. In the case of pluripotent stem cells, similar response mechanisms normally work and sometimes stimulate the differentiation cues. Up to now, differentiation protocols utilizing such environmental stresses have been reported to obtain various types of somatic cells from pluripotent stem cells. Basically, environmental stresses as hypoxia (low oxygen), hyperoxia, (high oxygen) and mechanical stress from cell culture plates are relatively safer than chemicals and gene transfers, which affect the genome irreversibly. Therefore, protocols designed with such environments in mind could be useful for the technology development of cell therapy and regenerative medicine. In this manuscript, we summarize recent findings of environmental stress-induced differentiation protocols and discuss their mechanisms. Abstract Pluripotent stem cells have unique characteristics compared to somatic cells. In this review, we summarize the response to environmental stresses (hypoxic, oxidative, thermal, and mechanical stresses) in embryonic stem cells (ESCs) and their applications in the differentiation methods directed to specific lineages. Those stresses lead to activation of each specific transcription factor followed by the induction of downstream genes, and one of them regulates lineage specification. In short, hypoxic stress promotes the differentiation of ESCs to mesodermal lineages via HIF-1α activation. Concerning mechanical stress, high stiffness tends to promote mesodermal differentiation, while low stiffness promotes ectodermal differentiation via the modulation of YAP1. Furthermore, each step in the same lineage differentiation favors each appropriate stiffness of culture plate; for example, definitive endoderm favors high stiffness, while pancreatic progenitor favors low stiffness during pancreatic differentiation of human ESCs. Overall, treatments utilizing those stresses have no genotoxic or carcinogenic effects except oxidative stress; therefore, the differentiated cells are safe and could be useful for cell replacement therapy. In particular, the effect of mechanical stress on differentiation is becoming attractive for the field of regenerative medicine. Therefore, the development of a stress-mediated differentiation protocol is an important matter for the future.
Collapse
|
16
|
Biomechanical Regulation of Stem Cell Fate. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-020-00183-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
17
|
Naqvi SM, McNamara LM. Stem Cell Mechanobiology and the Role of Biomaterials in Governing Mechanotransduction and Matrix Production for Tissue Regeneration. Front Bioeng Biotechnol 2020; 8:597661. [PMID: 33381498 PMCID: PMC7767888 DOI: 10.3389/fbioe.2020.597661] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Mechanobiology has underpinned many scientific advances in understanding how biophysical and biomechanical cues regulate cell behavior by identifying mechanosensitive proteins and specific signaling pathways within the cell that govern the production of proteins necessary for cell-based tissue regeneration. It is now evident that biophysical and biomechanical stimuli are as crucial for regulating stem cell behavior as biochemical stimuli. Despite this, the influence of the biophysical and biomechanical environment presented by biomaterials is less widely accounted for in stem cell-based tissue regeneration studies. This Review focuses on key studies in the field of stem cell mechanobiology, which have uncovered how matrix properties of biomaterial substrates and 3D scaffolds regulate stem cell migration, self-renewal, proliferation and differentiation, and activation of specific biological responses. First, we provide a primer of stem cell biology and mechanobiology in isolation. This is followed by a critical review of key experimental and computational studies, which have unveiled critical information regarding the importance of the biophysical and biomechanical cues for stem cell biology. This review aims to provide an informed understanding of the intrinsic role that physical and mechanical stimulation play in regulating stem cell behavior so that researchers may design strategies that recapitulate the critical cues and develop effective regenerative medicine approaches.
Collapse
Affiliation(s)
- S M Naqvi
- Mechanobiology and Medical Device Research Group, Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - L M McNamara
- Mechanobiology and Medical Device Research Group, Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
18
|
Zeevaert K, Elsafi Mabrouk MH, Wagner W, Goetzke R. Cell Mechanics in Embryoid Bodies. Cells 2020; 9:E2270. [PMID: 33050550 PMCID: PMC7599659 DOI: 10.3390/cells9102270] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Embryoid bodies (EBs) resemble self-organizing aggregates of pluripotent stem cells that recapitulate some aspects of early embryogenesis. Within few days, the cells undergo a transition from rather homogeneous epithelial-like pluripotent stem cell colonies into a three-dimensional organization of various cell types with multifaceted cell-cell interactions and lumen formation-a process associated with repetitive epithelial-mesenchymal transitions. In the last few years, culture methods have further evolved to better control EB size, growth, cellular composition, and organization-e.g., by the addition of morphogens or different extracellular matrix molecules. There is a growing perception that the mechanical properties, cell mechanics, and cell signaling during EB development are also influenced by physical cues to better guide lineage specification; substrate elasticity and topography are relevant, as well as shear stress and mechanical strain. Epithelial structures outside and inside EBs support the integrity of the cell aggregates and counteract mechanical stress. Furthermore, hydrogels can be used to better control the organization and lineage-specific differentiation of EBs. In this review, we summarize how EB formation is accompanied by a variety of biomechanical parameters that need to be considered for the directed and reproducible self-organization of early cell fate decisions.
Collapse
Affiliation(s)
- Kira Zeevaert
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Mohamed H. Elsafi Mabrouk
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Roman Goetzke
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, 52074 Aachen, Germany; (K.Z.); (M.H.E.M.)
- Institute for Biomedical Engineering–Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany
| |
Collapse
|
19
|
Evaluation of Dental Pulp Stem Cell Heterogeneity and Behaviour in 3D Type I Collagen Gels. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3034727. [PMID: 32964026 PMCID: PMC7501571 DOI: 10.1155/2020/3034727] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 01/09/2023]
Abstract
Dental pulp stem cells (DPSCs) are increasingly being advocated for regenerative medicine-based therapies. However, significant heterogeneity in the genotypic/phenotypic properties of DPSC subpopulations exist, influencing their therapeutic potentials. As most studies have established DPSC heterogeneity using 2D culture approaches, we investigated whether heterogeneous DPSC proliferative and contraction/remodelling capabilities were further evident within 3D type I collagen gels in vitro. DPSC subpopulations were isolated from human third molars and identified as high/low proliferative and multipotent/unipotent, following in vitro culture expansion and population doubling (PD) analysis. High proliferative/multipotent DPSCs, such as A3 (30 PDs and 80 PDs), and low proliferative/unipotent DPSCs, such as A1 (17 PDs), were cultured in collagen gels for 12 days, either attached or detached from the surrounding culture plastic. Collagen architecture and high proliferative/multipotent DPSC morphologies were visualised by Scanning Electron Microscopy and FITC-phalloidin/Fluorescence Microscopy. DPSC proliferation (cell counts), contraction (% diameter reductions), and remodelling (MMP-2/MMP-9 gelatin zymography) of collagen gels were also evaluated. Unexpectedly, no proliferation differences existed between DPSCs, A3 (30 PDs) and A1 (17 PDs), although A3 (80 PDs) responses were significantly reduced. Despite rapid detached collagen gel contraction with A3 (30 PDs), similar contraction rates were determined with A1 (17 PDs), although A3 (80 PDs) contraction was significantly impaired. Gel contraction correlated to distinct gelatinase profiles. A3 (30 PDs) possessed superior MMP-9 and comparable MMP-2 activities to A1 (17 PDs), whereas A3 (80 PDs) had significantly reduced MMP-2/MMP-9. High proliferative/multipotent DPSCs, A3 (30 PDs), further exhibited fibroblast-like morphologies becoming polygonal within attached gels, whilst losing cytoskeletal organization and fibroblastic morphologies in detached gels. This study demonstrates that heterogeneity exists in the gel contraction and MMP expression/activity capabilities of DPSCs, potentially reflecting differences in their abilities to degrade biomaterial scaffolds and regulate cellular functions in 3D environments and their regenerative properties overall. Thus, such findings enhance our understanding of the molecular and phenotypic characteristics associated with high proliferative/multipotent DPSCs.
Collapse
|
20
|
Hopf A, Schaefer DJ, Kalbermatten DF, Guzman R, Madduri S. Schwann Cell-Like Cells: Origin and Usability for Repair and Regeneration of the Peripheral and Central Nervous System. Cells 2020; 9:E1990. [PMID: 32872454 PMCID: PMC7565191 DOI: 10.3390/cells9091990] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
Functional recovery after neurotmesis, a complete transection of the nerve fiber, is often poor and requires a surgical procedure. Especially for longer gaps (>3 mm), end-to-end suturing of the proximal to the distal part is not possible, thus requiring nerve graft implantation. Artificial nerve grafts, i.e., hollow fibers, hydrogels, chitosan, collagen conduits, and decellularized scaffolds hold promise provided that these structures are populated with Schwann cells (SC) that are widely accepted to promote peripheral and spinal cord regeneration. However, these cells must be collected from the healthy peripheral nerves, resulting in significant time delay for treatment and undesired morbidities for the donors. Therefore, there is a clear need to explore the viable source of cells with a regenerative potential similar to SC. For this, we analyzed the literature for the generation of Schwann cell-like cells (SCLC) from stem cells of different origins (i.e., mesenchymal stem cells, pluripotent stem cells, and genetically programmed somatic cells) and compared their biological performance to promote axonal regeneration. Thus, the present review accounts for current developments in the field of SCLC differentiation, their applications in peripheral and central nervous system injury, and provides insights for future strategies.
Collapse
Affiliation(s)
- Alois Hopf
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
| | - Dirk J. Schaefer
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Daniel F. Kalbermatten
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Srinivas Madduri
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| |
Collapse
|
21
|
Zhang J, Huang YJ, Yoon JY, Kemmitt J, Wright C, Schneider K, Sphabmixay P, Hernandez-Gordillo V, Holcomb SJ, Bhushan B, Rohatgi G, Benton K, Carpenter D, Kester JC, Eng G, Breault DT, Yilmaz O, Taketani M, Voigt CA, Carrier RL, Trumper DL, Griffith LG. Primary human colonic mucosal barrier crosstalk with super oxygen-sensitive Faecalibacterium prausnitzii in continuous culture. MED 2020; 2:74-98.e9. [PMID: 33511375 DOI: 10.1016/j.medj.2020.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background The gut microbiome plays an important role in human health and disease. Gnotobiotic animal and in vitro cell-based models provide some informative insights into mechanistic crosstalk. However, there is no existing system for a long-term co-culture of a human colonic mucosal barrier with super oxygen-sensitive commensal microbes, hindering the study of human-microbe interactions in a controlled manner. Methods Here, we investigated the effects of an abundant super oxygen-sensitive commensal anaerobe, Faecalibacterium prausnitzii, on a primary human mucosal barrier using a Gut-MIcrobiome (GuMI) physiome platform that we designed and fabricated. Findings Long-term continuous co-culture of F. prausnitzii for two days with colon epithelia, enabled by continuous flow of completely anoxic apical media and aerobic basal media, resulted in a strictly anaerobic apical environment fostering growth of and butyrate production by F. prausnitzii, while maintaining a stable colon epithelial barrier. We identified elevated differentiation and hypoxia-responsive genes and pathways in the platform compared with conventional aerobic static culture of the colon epithelia, attributable to a combination of anaerobic environment and continuous medium replenishment. Furthermore, we demonstrated anti-inflammatory effects of F. prausnitzii through HDAC and the TLR-NFKB axis. Finally, we identified that butyrate largely contributes to the anti-inflammatory effects by downregulating TLR3 and TLR4. Conclusions Our results are consistent with some clinical observations regarding F. prausnitzii, thus motivating further studies employing this platform with more complex engineered colon tissues for understanding the interaction between the human colonic mucosal barrier and microbiota, pathogens, or engineered bacteria.
Collapse
Affiliation(s)
| | | | - Jun Young Yoon
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,School of Mechanical Engineering, Yonsei University, Seoul 03722, South Korea
| | | | | | | | | | | | | | - Brij Bhushan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gar Rohatgi
- EPAM Continuum, 41 University Drive, Newtown, PA 18940, USA
| | - Kyle Benton
- EPAM Continuum, 41 University Drive, Newtown, PA 18940, USA
| | | | | | | | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - David L Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda G Griffith
- Department of Biological Engineering.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
22
|
Davoodi E, Sarikhani E, Montazerian H, Ahadian S, Costantini M, Swieszkowski W, Willerth S, Walus K, Mofidfar M, Toyserkani E, Khademhosseini A, Ashammakhi N. Extrusion and Microfluidic-based Bioprinting to Fabricate Biomimetic Tissues and Organs. ADVANCED MATERIALS TECHNOLOGIES 2020; 5:1901044. [PMID: 33072855 PMCID: PMC7567134 DOI: 10.1002/admt.201901044] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/10/2020] [Indexed: 05/07/2023]
Abstract
Next generation engineered tissue constructs with complex and ordered architectures aim to better mimic the native tissue structures, largely due to advances in three-dimensional (3D) bioprinting techniques. Extrusion bioprinting has drawn tremendous attention due to its widespread availability, cost-effectiveness, simplicity, and its facile and rapid processing. However, poor printing resolution and low speed have limited its fidelity and clinical implementation. To circumvent the downsides associated with extrusion printing, microfluidic technologies are increasingly being implemented in 3D bioprinting for engineering living constructs. These technologies enable biofabrication of heterogeneous biomimetic structures made of different types of cells, biomaterials, and biomolecules. Microfluiding bioprinting technology enables highly controlled fabrication of 3D constructs in high resolutions and it has been shown to be useful for building tubular structures and vascularized constructs, which may promote the survival and integration of implanted engineered tissues. Although this field is currently in its early development and the number of bioprinted implants is limited, it is envisioned that it will have a major impact on the production of customized clinical-grade tissue constructs. Further studies are, however, needed to fully demonstrate the effectiveness of the technology in the lab and its translation to the clinic.
Collapse
Affiliation(s)
- Elham Davoodi
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Einollah Sarikhani
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Hossein Montazerian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Marco Costantini
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, 00-661 Warsaw, Poland
- Institute of Physical Chemistry – Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, 00-661 Warsaw, Poland
| | - Stephanie Willerth
- Department of Mechanical Engineering, Division of Medical Sciences, University of Victoria, BC V8P 5C2, Canada
| | - Konrad Walus
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mohammad Mofidfar
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Ehsan Toyserkani
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
23
|
Lee HR, Sung JH. Effect of culture condition on cell viability and gel contraction in a skin chip. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Gonzalez BA, Perez-Nevarez M, Mirza A, Perez MG, Lin YM, Hsu CPD, Caobi A, Raymond A, Gomez Hernandez ME, Fernandez-Lima F, George F, Ramaswamy S. Physiologically Relevant Fluid-Induced Oscillatory Shear Stress Stimulation of Mesenchymal Stem Cells Enhances the Engineered Valve Matrix Phenotype. Front Cardiovasc Med 2020; 7:69. [PMID: 32509802 PMCID: PMC7248568 DOI: 10.3389/fcvm.2020.00069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/07/2020] [Indexed: 11/20/2022] Open
Abstract
Support of somatic growth is a fundamental requirement of tissue-engineered valves. However, efforts thus far have been unable to maintain this support long term. A key event that will determine the valve's long-term success is the extent to which healthy host tissue remodeling can occur on the valve soon after implantation. The construct's phenotypic-status plays a critical role in accelerating tissue remodeling and engineered valve integration with the host via chemotaxis. In the current study, human bone-marrow-derived mesenchymal stem cells were utilized to seed synthetic, biodegradable scaffolds for a period of 8 days in rotisserie culture. Subsequently, cell-seeded scaffolds were exposed to physiologically relevant oscillatory shear stresses (overall mean, time-averaged shear stress, ~7.9 dynes/cm2; overall mean, oscillatory shear index, ~0.18) for an additional 2 weeks. The constructs were found to exhibit relatively augmented endothelial cell expression (CD31; compared to static controls) but concomitantly served to restrict the level of the activated smooth muscle phenotype (α-SMA) and also produced very low stem cell secretion levels of fibronectin (p < 0.05 compared to static and rotisserie controls). These findings suggest that fluid-induced oscillatory shear stresses alone are important in regulating a healthy valve phenotype of the engineered tissue matrix. Moreover, as solid stresses could lead to increased α-SMA levels, they should be excluded from conditioning during the culture process owing to their associated potential risks with pathological tissue remodeling. In conclusion, engineered valve tissues derived from mesenchymal stem cells revealed both a relatively robust valvular phenotype after exposure to physiologically relevant scales of oscillatory shear stress and may thereby serve to accelerate healthy valve tissue remodeling in the host post-implantation.
Collapse
Affiliation(s)
- Brittany A Gonzalez
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Manuel Perez-Nevarez
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Asad Mirza
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Marcos Gonzalez Perez
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Yih-Mei Lin
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Chia-Pei Denise Hsu
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Allen Caobi
- Department of Immunology and Nano-Medicine, Florida International University, Miami, FL, United States
| | - Andrea Raymond
- Department of Immunology and Nano-Medicine, Florida International University, Miami, FL, United States
| | - Mario E Gomez Hernandez
- Advanced Mass Spectrometry Facility, Florida International University, Miami, FL, United States.,Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States
| | - Francisco Fernandez-Lima
- Advanced Mass Spectrometry Facility, Florida International University, Miami, FL, United States.,Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States
| | - Florence George
- Department of Mathematics and Statistics, Florida International University, Miami, FL, United States
| | - Sharan Ramaswamy
- Cardiovascular Therapeutics Laboratory (CV-PEUTICS Lab), Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| |
Collapse
|
25
|
Vander Roest MJ, Merryman WD. Cyclic Strain Promotes H19 Expression and Vascular Tube Formation in iPSC-Derived Endothelial Cells. Cell Mol Bioeng 2020; 13:369-377. [PMID: 32952736 DOI: 10.1007/s12195-020-00617-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Induced pluripotent stem cell (iPSC)-derived endothelial cells (ECs) have the potential for therapeutic application in several cardiovascular diseases. Mechanical strain is known to regulate EC behavior and stem cell differentiation and may play a role in directing EC differentiation of iPSCs. H19, a long non-coding RNA (lncRNA), is known to affect ECs in several mechanically relevant pathologies and may play a role in this process as well. Therefore, we investigated expression changes of H19 resulting from mechanical stimulation during EC differentiation, as well as functional effects on EC tube formation. Methods iPSCs were subjected to 5% cyclic mechanical strain during EC differentiation. RT-PCR and flow cytometry were used to assess changes in mesoderm differentiation and gene expression in the final ECs as a result of strain. Functional outcomes of mechanically differentiated ECs were assessed with a tube formation assay and changes in H19. H19 was also overexpressed in human umbilical vein endothelial cells (HUVECs) to assess its role in non-H19-expressing ECs. Results Mechanical strain promoted mesoderm differentiation, marked by increased expression of brachyury 24 h after initiation of differentiation. Strain also increased expression of H19, CD31, VE-cadherin, and VEGFR2 in differentiated ECs. Strain-differentiated ECs formed tube networks with higher junction and endpoint density than statically-differentiated ECs. Overexpression of H19 in HUVECs resulted in similar patterns of tube formation. Conclusions H19 expression is increased by mechanical strain and promotes tube branching in iPSC-derived ECs.
Collapse
Affiliation(s)
- Mark J Vander Roest
- Biomedical Engineering, Vanderbilt University, Room 9445D MRB4, 2213 Garland Ave, Nashville, TN 37212 USA
| | - W David Merryman
- Biomedical Engineering, Vanderbilt University, Room 9445D MRB4, 2213 Garland Ave, Nashville, TN 37212 USA
| |
Collapse
|
26
|
Yumoto M, Hemmi N, Sato N, Kawashima Y, Arikawa K, Ide K, Hosokawa M, Seo M, Takeyama H. Evaluation of the effects of cell-dispensing using an inkjet-based bioprinter on cell integrity by RNA-seq analysis. Sci Rep 2020; 10:7158. [PMID: 32346113 PMCID: PMC7189371 DOI: 10.1038/s41598-020-64193-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/10/2020] [Indexed: 12/22/2022] Open
Abstract
Bioprinting technology is expected to be applied in the fields of regenerative medicine and drug discovery. There are several types of bioprinters, especially inkjet-based bioprinter, which can be used not only as a printer for arranging cells but also as a precision cell-dispensing device with controlled cell numbers similar to a fluorescence activated cell sorter (FACS). Precise cell dispensers are expected to be useful in the fields of drug discovery and single-cell analysis. However, there are enduring concerns about the impacts of cell dispensers on cell integrity, particularly on sensitive cells, such as stem cells. In response to the concerns stated above, we developed a stress-free and media-direct-dispensing inkjet bioprinter. In the present study, in addition to conventional viability assessments, we evaluated the gene expression using RNA-seq to investigate whether the developed bioprinter influenced cell integrity in mouse embryonic stem cells. We evaluated the developed bioprinter based on three dispensing methods: manual operation using a micropipette, FACS and the developed inkjet bioprinter. According to the results, the developed inkjet bioprinter exhibited cell-friendly dispensing performance, which was similar to the manual dispensing operation, based not only on cell viability but also on gene expression levels.
Collapse
Affiliation(s)
- Masayuki Yumoto
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Biomedical Business Center, Healthcare Business Group, Ricoh Company, Ltd., 3-25-22 Tonomachi LIC 322, Kawasaki, Kanagawa, 210-0821, Japan
| | - Natsuko Hemmi
- Biomedical Business Center, Healthcare Business Group, Ricoh Company, Ltd., 3-25-22 Tonomachi LIC 322, Kawasaki, Kanagawa, 210-0821, Japan
| | - Naoki Sato
- Biomedical Business Center, Healthcare Business Group, Ricoh Company, Ltd., 3-25-22 Tonomachi LIC 322, Kawasaki, Kanagawa, 210-0821, Japan
| | - Yudai Kawashima
- Biomedical Business Center, Healthcare Business Group, Ricoh Company, Ltd., 3-25-22 Tonomachi LIC 322, Kawasaki, Kanagawa, 210-0821, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda University, 513 Waseda-tsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Keigo Ide
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Computational Bio Big-Data Open Innovation Laboratory, AIST-Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Masahito Hosokawa
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
- Research Organization for Nano and Life Innovation, Waseda University, 513 Waseda-tsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Manabu Seo
- Biomedical Business Center, Healthcare Business Group, Ricoh Company, Ltd., 3-25-22 Tonomachi LIC 322, Kawasaki, Kanagawa, 210-0821, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan.
- Computational Bio Big-Data Open Innovation Laboratory, AIST-Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
- Research Organization for Nano and Life Innovation, Waseda University, 513 Waseda-tsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan.
| |
Collapse
|
27
|
Pearce HA, Kim YS, Diaz-Gomez L, Mikos AG. Tissue Engineering Scaffolds. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00082-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
28
|
Choi JH, Santhosh M, Choi JW. In Vitro Blood-Brain Barrier-Integrated Neurological Disorder Models Using a Microfluidic Device. MICROMACHINES 2019; 11:E21. [PMID: 31878184 PMCID: PMC7019695 DOI: 10.3390/mi11010021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) plays critical role in the human physiological system such as protection of the central nervous system (CNS) from external materials in the blood vessel, including toxicants and drugs for several neurological disorders, a critical type of human disease. Therefore, suitable in vitro BBB models with fluidic flow to mimic the shear stress and supply of nutrients have been developed. Neurological disorder has also been investigated for developing realistic models that allow advance fundamental and translational research and effective therapeutic strategy design. Here, we discuss introduction of the blood-brain barrier in neurological disorder models by leveraging a recently developed microfluidic system and human organ-on-a-chip system. Such models could provide an effective drug screening platform and facilitate personalized therapy of several neurological diseases.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
| | - Mallesh Santhosh
- Center for Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
- Center for Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
| |
Collapse
|
29
|
Urbanczyk M, Layland SL, Schenke-Layland K. The role of extracellular matrix in biomechanics and its impact on bioengineering of cells and 3D tissues. Matrix Biol 2019; 85-86:1-14. [PMID: 31805360 DOI: 10.1016/j.matbio.2019.11.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/24/2019] [Accepted: 11/24/2019] [Indexed: 12/20/2022]
Abstract
The cells and tissues of the human body are constantly exposed to exogenous and endogenous forces that are referred to as biomechanical cues. They guide and impact cellular processes and cell fate decisions on the nano-, micro- and macro-scale, and are therefore critical for normal tissue development and maintaining tissue homeostasis. Alterations in the extracellular matrix composition of a tissue combined with abnormal mechanosensing and mechanotransduction can aberrantly activate signaling pathways that promote disease development. Such processes are therefore highly relevant for disease modelling or when aiming for the development of novel therapies. In this mini review, we describe the main biomechanical cues that impact cellular fates. We highlight their role during development, homeostasis and in disease. We also discuss current techniques and tools that allow us to study the impact of biomechanical cues on cell and tissue development under physiological conditions, and we point out directions, in which in vitro biomechanics can be of use in the future.
Collapse
Affiliation(s)
- Max Urbanczyk
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Germany
| | - Shannon L Layland
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Germany; Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany; Cluster of Excellence IFIT (EXC 2180), "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Germany; Dept. of Medicine/Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
30
|
Deng C, He Y, Feng J, Dong Z, Yao Y, Lu F. Conditioned medium from 3D culture system of stromal vascular fraction cells accelerates wound healing in diabetic rats. Regen Med 2019; 14:925-937. [PMID: 31599183 DOI: 10.2217/rme-2018-0083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aim: We investigated the healing effects of conditioned medium (CM) derived from a physiological 3D culture system engineered to use an extracellular matrix/stromal vascular fraction (SVF) gel enriched for adipose on diabetic wounds in rats. This CM (Gel-CM) was compared with that from a 2D culture system that used SVF cells (SVF-CM). Materials & methods: Keratinocytes, fibroblasts and wounds were treated with Gel-CM and SVF-CM, and cytokine levels in the CM types were quantified. Results: Proliferation and migration of keratinocytes and fibroblasts were significantly higher after treatment with Gel-CM than with SVF-CM. Collagen secretion by fibroblasts and wound closure were highly stimulated by Gel-CM. Proteomic analyses revealed a higher concentration of growth factors in Gel-CM than in SVF-CM. Conclusion: Gel-CM is a promising therapeutic option for treating diabetic wounds.
Collapse
Affiliation(s)
- Chengliang Deng
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563000, PR China.,Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yunfan He
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Ziqing Dong
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yao Yao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
31
|
Li R, Baek KI, Chang CC, Zhou B, Hsiai TK. Mechanosensitive Pathways Involved in Cardiovascular Development and Homeostasis in Zebrafish. J Vasc Res 2019; 56:273-283. [PMID: 31466069 DOI: 10.1159/000501883] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 07/03/2019] [Indexed: 11/19/2022] Open
Abstract
Cardiovascular diseases such as coronary heart disease, myocardial infarction, and cardiac arrhythmia are the leading causes of morbidity and mortality in developed countries and are steadily increasing in developing countries. Fundamental mechanistic studies at the molecular, cellular, and animal model levels are critical for the diagnosis and treatment of these diseases. Despite being phylogenetically distant from humans, zebrafish share remarkable similarity in the genetics and electrophysiology of the cardiovascular system. In the last 2 decades, the development and deployment of innovative genetic manipulation techniques greatly facilitated the application of zebrafish as an animal model for studying basic biology and diseases. Hemodynamic shear stress is intimately involved in vascular development and homeostasis. The critical mechanosensitive signaling pathways in cardiovascular development and pathophysiology previously studied in mammals have been recapitulated in zebrafish. In this short article, we reviewed recent knowledge about the role of mechanosensitive pathways such as Notch, PKCε/PFKFB3, and Wnt/Ang2 in cardiovas-cular development and homeostasis from studies in the -zebrafish model.
Collapse
Affiliation(s)
- Rongsong Li
- College of Health Sciences and Environmental Engineering, Shenzhen Technology University, Shenzhen, China,
| | - Kyung In Baek
- Department of Bioengineering,University of California, Los Angeles, California, USA
| | - Chih-Chiang Chang
- Department of Bioengineering,University of California, Los Angeles, California, USA
| | - Bill Zhou
- Department of Radiology, University of California, Los Angeles, California, USA
| | - Tzung K Hsiai
- Department of Bioengineering,University of California, Los Angeles, California, USA.,Department of Medicine (Cardiology) and Bioengineering, University of California, Los Angeles, California, USA
| |
Collapse
|
32
|
Microfluidic models of physiological or pathological flow shear stress for cell biology, disease modeling and drug development. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Sonnen KF, Merten CA. Microfluidics as an Emerging Precision Tool in Developmental Biology. Dev Cell 2019; 48:293-311. [PMID: 30753835 DOI: 10.1016/j.devcel.2019.01.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/13/2018] [Accepted: 01/10/2019] [Indexed: 12/18/2022]
Abstract
Microfluidics has become a precision tool in modern biology. It enables omics data to be obtained from individual cells, as compared to averaged signals from cell populations, and it allows manipulation of biological specimens in entirely new ways. Cells and organisms can be perturbed at extraordinary spatiotemporal resolution, revealing mechanistic insights that would otherwise remain hidden. In this perspective article, we discuss the current and future impact of microfluidic technology in the field of developmental biology. In addition, we provide detailed information on how to start using this technology even without prior experience.
Collapse
Affiliation(s)
| | - Christoph A Merten
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
34
|
Gopal S, Chiappini C, Armstrong JPK, Chen Q, Serio A, Hsu CC, Meinert C, Klein TJ, Hutmacher DW, Rothery S, Stevens MM. Immunogold FIB-SEM: Combining Volumetric Ultrastructure Visualization with 3D Biomolecular Analysis to Dissect Cell-Environment Interactions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1900488. [PMID: 31197896 PMCID: PMC6778054 DOI: 10.1002/adma.201900488] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/20/2019] [Indexed: 05/03/2023]
Abstract
Volumetric imaging techniques capable of correlating structural and functional information with nanoscale resolution are necessary to broaden the insight into cellular processes within complex biological systems. The recent emergence of focused ion beam scanning electron microscopy (FIB-SEM) has provided unparalleled insight through the volumetric investigation of ultrastructure; however, it does not provide biomolecular information at equivalent resolution. Here, immunogold FIB-SEM, which combines antigen labeling with in situ FIB-SEM imaging, is developed in order to spatially map ultrastructural and biomolecular information simultaneously. This method is applied to investigate two different cell-material systems: the localization of histone epigenetic modifications in neural stem cells cultured on microstructured substrates and the distribution of nuclear pore complexes in myoblasts differentiated on a soft hydrogel surface. Immunogold FIB-SEM offers the potential for broad applicability to correlate structure and function with nanoscale resolution when addressing questions across cell biology, biomaterials, and regenerative medicine.
Collapse
Affiliation(s)
- Sahana Gopal
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Ciro Chiappini
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - James P K Armstrong
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Qu Chen
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Andrea Serio
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Chia-Chen Hsu
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Christoph Meinert
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
| | - Travis J Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
- Australian Research Council Industrial Transformation Training Centre, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
- Australian Research Council Industrial Transformation Training Centre, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
| | - Stephen Rothery
- Facility for Light Microscopy, Imperial College London, London, SW7 2AZ, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
35
|
Moosazadeh Moghaddam M, Bonakdar S, Shokrgozar MA, Zaminy A, Vali H, Faghihi S. Engineered substrates with imprinted cell-like topographies induce direct differentiation of adipose-derived mesenchymal stem cells into Schwann cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1022-1035. [PMID: 30942113 DOI: 10.1080/21691401.2019.1586718] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Differentiation of stem cells to Schwann is considered efficient way for nerve regeneration since the sources of human Schwann cells are limited for clinical application. It is demonstrated that mimicking micromechanical forces or micro/nanotopographical environments that stem cells are experienced in vivo could control their fate. Here, the potency of substrates with imprinted cell-like topographies for direct differentiation of adipose-derived mesenchymal stem cells (ADSCs) into Schwann cells (SCs) is reported. For the preparation of substrates with imprinted SC-Like topographies, SCs are isolated from the sciatic nerve, grown, fixed, and then SC morphologies are transferred to polydimethylsiloxane (PDMS) substrates by mold casting. Subsequently, mesenchymal stem cells (MSCs) are seeded on the SC-imprinted substrates and their differentiation to SCs is evaluated by immunocytochemistry, real-time PCR, and western blotting. Analysis of morphology and expression of SC-specific markers show that MSCs cultured on the imprinted substrates have the typical SC-like morphology and express SC-specific markers including S100b, p75NTR, and Sox10. It is believed that specific cell-like topographies and related micromechanical cues can be sufficient for direct differentiation of ADSCs into Schwann cells by cell-imprinting method as a physical technique.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- a Stem Cell and Regenerative Medicine Group , National Institute of Genetic Engineering and Biotechnology (NIGEB) , Tehran , Iran
| | - Shahin Bonakdar
- b National Cell Bank , Pasteur Institute of Iran , Tehran , Iran
| | | | - Arash Zaminy
- c Neuroscience Research Center, Faculty of Medicine , Guilan University of Medical Sciences , Rasht , Iran
| | - Hojatollah Vali
- d Department of Anatomy and Cell Biology , McGill University , Montréal , QC , Canada.,e Facility for Electron Microscopy Research , McGill University , Montréal , QC , Canada
| | - Shahab Faghihi
- a Stem Cell and Regenerative Medicine Group , National Institute of Genetic Engineering and Biotechnology (NIGEB) , Tehran , Iran
| |
Collapse
|
36
|
Moghaddam MM, Bonakdar S, Shariatpanahi MR, Shokrgozar MA, Faghihi S. The Effect of Physical Cues on the Stem Cell Differentiation. Curr Stem Cell Res Ther 2019; 14:268-277. [DOI: 10.2174/1574888x14666181227120706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/05/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022]
Abstract
Development of multicellular organisms is a very complex and organized process during which cells respond to various factors and features in extracellular environments. It has been demonstrated that during embryonic evolvement, under certain physiological or experimental conditions, unspecialized cells or stem cells can be induced to become tissue or organ-specific cells with special functions. Considering the importance of physical cues in stem cell fate, the present study reviews the role of physical factors in stem cells differentiation and discusses the molecular mechanisms associated with these factors.
Collapse
Affiliation(s)
- Mehrdad M. Moghaddam
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, 14965/161, Iran
| | - Shahin Bonakdar
- National Cell Bank, Pasteur Institute of Iran, Tehran 3159915111, Iran
| | | | | | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, 14965/161, Iran
| |
Collapse
|
37
|
Low-level shear stress induces differentiation of liver cancer stem cells via the Wnt/β-catenin signalling pathway. Exp Cell Res 2019; 375:90-96. [DOI: 10.1016/j.yexcr.2018.12.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/24/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022]
|
38
|
Akutagawa T, Aoki S, Yamamoto-Rikitake M, Iwakiri R, Fujimoto K, Toda S. Cancer-adipose tissue interaction and fluid flow synergistically modulate cell kinetics, HER2 expression, and trastuzumab efficacy in gastric cancer. Gastric Cancer 2018; 21:946-955. [PMID: 29696406 DOI: 10.1007/s10120-018-0829-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Early local tumor invasion in gastric cancer results in likely encounters between cancer cells and submucosal and subserosal adipose tissue, but these interactions remain to be clarified. Microenvironmental mechanical forces, such as fluid flow, are known to modulate normal cell kinetics, but the effects of fluid flow on gastric cancer cells are poorly understood. We analyzed the cell kinetics and chemosensitivity in gastric cancer using a simple in vitro model that simultaneously replicated the cancer-adipocyte interaction and physical microenvironment. METHODS Gastric cancer cells (MKN7 and MKN74) were seeded on rat adipose tissue fragment-embedded discs or collagen discs alone. To generate fluid flow, samples were placed on a rotatory shaker in a CO2 incubator. Proliferation, apoptosis, invasion, and motility-related molecules were analyzed by morphometry and immunostaining. Proteins were evaluated by western blot analysis. Chemosensitivity was investigated by trastuzumab treatment. RESULTS Adipose tissue and fluid flow had a positive synergistic effect on the proliferative potential and invasive capacity of gastric cancer cells, and adipose tissue inhibited apoptosis in these cells. Adipose tissue upregulated ERK1/2 signaling in gastric cancer cells, but downregulated p38 signaling. Notably, adipose tissue and fluid flow promoted membranous and cytoplasmic HER2 expression and modulated chemosensitivity to trastuzumab in gastric cancer cells. CONCLUSION We have demonstrated that cancer-adipocyte interaction and physical microenvironment mutually modulate gastric cancer cell kinetics. Further elucidation of the microenvironmental regulation in gastric cancer will be very important for the development of strategies involving molecular targeted therapy.
Collapse
Affiliation(s)
- Takashi Akutagawa
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan. .,Department of Internal Medicine and Gastrointestinal Endoscopy, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan.
| | - Shigehisa Aoki
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan.
| | - Mihoko Yamamoto-Rikitake
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Ryuichi Iwakiri
- Department of Internal Medicine and Gastrointestinal Endoscopy, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Kazuma Fujimoto
- Department of Internal Medicine and Gastrointestinal Endoscopy, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| | - Shuji Toda
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, Saga, 849-8501, Japan
| |
Collapse
|
39
|
Mashimo Y, Yoshioka M, Tokunaga Y, Fockenberg C, Terada S, Koyama Y, Shibata-Seki T, Yoshimoto K, Sakai R, Hakariya H, Liu L, Akaike T, Kobatake E, How SE, Uesugi M, Chen Y, Kamei KI. Fabrication of a Multiplexed Artificial Cellular MicroEnvironment Array. J Vis Exp 2018. [PMID: 30247461 DOI: 10.3791/57377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cellular microenvironments consist of a variety of cues, such as growth factors, extracellular matrices, and intercellular interactions. These cues are well orchestrated and are crucial in regulating cell functions in a living system. Although a number of researchers have attempted to investigate the correlation between environmental factors and desired cellular functions, much remains unknown. This is largely due to the lack of a proper methodology to mimic such environmental cues in vitro, and simultaneously test different environmental cues on cells. Here, we report an integrated platform of microfluidic channels and a nanofiber array, followed by high-content single-cell analysis, to examine stem cell phenotypes altered by distinct environmental factors. To demonstrate the application of this platform, this study focuses on the phenotypes of self-renewing human pluripotent stem cells (hPSCs). Here, we present the preparation procedures for a nanofiber array and the microfluidic structure in the fabrication of a Multiplexed Artificial Cellular MicroEnvironment (MACME) array. Moreover, overall steps of the single-cell profiling, cell staining with multiple fluorescent markers, multiple fluorescence imaging, and statistical analyses, are described.
Collapse
Affiliation(s)
- Yasumasa Mashimo
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology
| | - Momoko Yoshioka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Yumie Tokunaga
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | | | - Shiho Terada
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Yoshie Koyama
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Teiko Shibata-Seki
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology
| | - Koki Yoshimoto
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Risako Sakai
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Hayase Hakariya
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Li Liu
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Toshihiro Akaike
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science
| | - Eiry Kobatake
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology
| | - Siew-Eng How
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah
| | - Motonari Uesugi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University; Institute for Chemical Research, Kyoto University
| | - Yong Chen
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University; Ecole Normale Supérieure
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University;
| |
Collapse
|
40
|
Dorsey TB, Kim D, Grath A, James D, Dai G. Multivalent biomaterial platform to control the distinct arterial venous differentiation of pluripotent stem cells. Biomaterials 2018; 185:1-12. [PMID: 30216805 DOI: 10.1016/j.biomaterials.2018.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 11/25/2022]
Abstract
Vascular endothelial cells (ECs) differentiated from pluripotent stem cells have enormous potential to be used in a variety of therapeutic areas such as tissue engineering of vascular grafts and re-vascularization of ischemic tissues. To date, various protocols have been developed to differentiate stem cells toward vascular ECs. However, current methods are still not sufficient to drive the distinct arterial venous differentiation. Therefore, developing refined method of arterial-venous differentiation is critically needed to address this gap. Here, we developed a biomaterial platform to mimic multivalent ephrin-B2/EphB4 signaling and investigated its role in the early arterial and venous specification of pluripotent stem cells. Our results show immobilized ephrinB2 or EphB4 on hydrogel substrates have a distinct effect on arterial venous differentiation by regulating several arterial venous markers. When in combination with Wnt pathway agonist or BMP4 signaling, the ephrin-B2/EphB4 biomaterial platform can create diverging EC progenitor populations, demonstrating differential gene expression pattern across a wide range of arterial and venous markers, as well as phenotypic markers such as anti-thrombotic, pro-atherogenic and osteogenic genes, that are consistent with the in vivo expression patterns of arterial and venous ECs. Importantly, this distinct EC progenitor population cannot be achieved by current methods of applying soluble factors or hemodynamic stimuli alone, illustrating that fine-tuning of developmental signals using the biomaterial platform offers a new approach to better control the arterial venous differentiation of stem cells.
Collapse
Affiliation(s)
- Taylor B Dorsey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Diana Kim
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Alexander Grath
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Daylon James
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10065, United States
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States.
| |
Collapse
|
41
|
Ort C, Dayekh K, Xing M, Mequanint K. Emerging Strategies for Stem Cell Lineage Commitment in Tissue Engineering and Regenerative Medicine. ACS Biomater Sci Eng 2018; 4:3644-3657. [PMID: 33429592 DOI: 10.1021/acsbiomaterials.8b00532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stem cells have transformed the fields of tissue engineering and regenerative medicine, and their potential to further advance these fields cannot be overstated. The stem cell niche is a dynamic microenvironment that determines cell fate during development and tissue repair following an injury. Classically, stem cells were studied in isolation of their microenvironment; however, contemporary research has produced a myriad of evidence that shows the importance of multiple aspects of the stem cell niche in regulating their processes. In the context of tissue engineering and regenerative medicine studies, the niche is an artificial environment provided by culture conditions. In vitro culture conditions may involve coculturing with other cell types, developing specific biomaterials, and applying relevant forces to promote the desired lineage commitment. Considerable advance has been made over the past few years toward directed stem cell differentiation; however, the unspecific differentiation of stem cells yielding a mixed population of cells has been a challenge. In this review, we provide a systematic review of the emerging strategies used for lineage commitment within the context of tissue engineering and regenerative medicine. These strategies include scaffold pore-size and pore-shape gradients, stress relaxation, sonic and electromagnetic effects, and magnetic forces. Finally, we provide insights and perspectives into future directions focusing on signaling pathways activated during lineage commitment using external stimuli.
Collapse
Affiliation(s)
| | | | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, 66 Chancellors Circle, Winnipeg R3T 2N2, Canada
| | | |
Collapse
|
42
|
Abstract
Vascular, resident stem cells are present in all 3 layers of the vessel wall; they play a role in vascular formation under physiological conditions and in remodeling in pathological situations. Throughout development and adult early life, resident stem cells participate in vessel formation through vasculogenesis and angiogenesis. In adults, the vascular stem cells are mostly quiescent in their niches but can be activated in response to injury and participate in endothelial repair and smooth muscle cell accumulation to form neointima. However, delineation of the characteristics and of the migration and differentiation behaviors of these stem cells is an area of ongoing investigation. A set of genetic mouse models for cell lineage tracing has been developed to specifically address the nature of these cells and both migration and differentiation processes during physiological angiogenesis and in vascular diseases. This review summarizes the current knowledge on resident stem cells, which has become more defined and refined in vascular biology research, thus contributing to the development of new potential therapeutic strategies to promote endothelial regeneration and ameliorate vascular disease development.
Collapse
Affiliation(s)
- Li Zhang
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Shirin Issa Bhaloo
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| | - Ting Chen
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences (B.Z.)
| | - Qingbo Xu
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| |
Collapse
|
43
|
Yamamoto T, Yano M, Okano Y, Kino-oka M. Numerical Investigation for the Movement of Cell Colonies in Bioreactors: Stirring and Orbital Shaking Tanks. JOURNAL OF CHEMICAL ENGINEERING OF JAPAN 2018. [DOI: 10.1252/jcej.17we112] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Takuya Yamamoto
- Department of Materials Engineering Science, Osaka University
| | - Masaki Yano
- Department of Materials Engineering Science, Osaka University
| | - Yasunori Okano
- Department of Materials Engineering Science, Osaka University
| | | |
Collapse
|
44
|
Kim D, Lee V, Dorsey TB, Niklason LE, Gui L, Dai G. Neuropilin-1 Mediated Arterial Differentiation of Murine Pluripotent Stem Cells. Stem Cells Dev 2018; 27:441-455. [PMID: 29415620 DOI: 10.1089/scd.2017.0240] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Pluripotent stem cell-derived endothelial cells (ECs) have great potential to be used in vascular therapy or tissue engineering. It is also much desired to obtain arterial or venous ECs for specific applications. Factors that are critical for the proper arterial or venous differentiation from pluripotent stem cells still need to be understood. Here, we aim at investigating this problem deeper by examining neuropilin-1 (Nrp1), an early arterial marker that may be critical for arterial cell fate commitment. Using murine embryonic stem cells as the model system, this study investigates the neuropilin-1 (Nrp1) expression during the differentiation of pluripotent stem cells toward a vascular progenitor population. We hypothesize that Nrp1, an early arterial marker present in a developing embryo, may be more responsive when further induced in vitro toward an arterial fate. We developed a two-step differentiation approach that yielded a large percentage of Nrp1+ vascular progenitor cells (VPCs) and investigated their potential to become arterial ECs. We have defined the culture parameters that contribute greatly to the emergence of Nrp1+ VPCs: certain soluble factors, especially Wnt and BMP4, early cell-cell contact, and hypoxia. Subsequent isolation of this population demonstrated a highly proliferative and network-forming behavior. The Nrp1+ VPCs exhibited increased gene expression of several Notch pathway-related arterial markers compared with Nrp1- VPCs. Most importantly, Nrp1+ VPCs demonstrated a dramatically greater response to hemodynamic stimuli by upregulating many arterial markers whereas Nrp1- VPCs have very little response. Surprisingly, these differences between Nrp1+ and Nrp1- VPCs are not evident with vascular endothelial growth factor (VEGF) treatment. Our data suggest that Nrp1+ VPCs may serve as the arterial progenitor by enhanced response to hemodynamic flow but not to VEGF, whereas Nrp1- VPCs lack the plasticity to become arterial ECs. The findings of this research indicate that Nrp1+ VPCs in the murine model act as an important step in the arterial differentiation process.
Collapse
Affiliation(s)
- Diana Kim
- 1 Department of Biomedical Engineering, Rensselaer Polytechnic Institute , Troy, New York.,2 Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York.,3 Department of Bioengineering, Northeastern University , Boston, Massachusetts
| | - Vivian Lee
- 1 Department of Biomedical Engineering, Rensselaer Polytechnic Institute , Troy, New York.,2 Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York.,3 Department of Bioengineering, Northeastern University , Boston, Massachusetts
| | - Taylor B Dorsey
- 1 Department of Biomedical Engineering, Rensselaer Polytechnic Institute , Troy, New York.,2 Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York.,3 Department of Bioengineering, Northeastern University , Boston, Massachusetts
| | - Laura E Niklason
- 4 Vascular Biology and Therapeutics Program, Yale University School of Medicine , New Haven, Connecticut.,5 Department of Anesthesiology, Yale University , New Haven, Connecticut.,6 Department of Biomedical Engineering, Yale University , New Haven, Connecticut.,7 Yale Stem Cell Center , New Haven, Connecticut
| | - Liqiong Gui
- 4 Vascular Biology and Therapeutics Program, Yale University School of Medicine , New Haven, Connecticut.,5 Department of Anesthesiology, Yale University , New Haven, Connecticut
| | - Guohao Dai
- 1 Department of Biomedical Engineering, Rensselaer Polytechnic Institute , Troy, New York.,2 Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York.,3 Department of Bioengineering, Northeastern University , Boston, Massachusetts
| |
Collapse
|
45
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
46
|
La A, Tranquillo RT. Hemocompatible tissue-engineered vascular grafts using adult mesenchymal stem cells. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Whole-Transcriptome Sequencing: a Powerful Tool for Vascular Tissue Engineering and Endothelial Mechanobiology. High Throughput 2018; 7:ht7010005. [PMID: 29485616 PMCID: PMC5876531 DOI: 10.3390/ht7010005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023] Open
Abstract
Among applicable high-throughput techniques in cardiovascular biology, whole-transcriptome sequencing is of particular use. By utilizing RNA that is isolated from virtually all cells and tissues, the entire transcriptome can be evaluated. In comparison with other high-throughput approaches, RNA sequencing is characterized by a relatively low-cost and large data output, which permits a comprehensive analysis of spatiotemporal variation in the gene expression profile. Both shear stress and cyclic strain exert hemodynamic force upon the arterial endothelium and are considered to be crucial determinants of endothelial physiology. Laminar blood flow results in a high shear stress that promotes atheroresistant endothelial phenotype, while a turbulent, oscillatory flow yields a pathologically low shear stress that disturbs endothelial homeostasis, making respective arterial segments prone to atherosclerosis. Severe atherosclerosis significantly impairs blood supply to the organs and frequently requires bypass surgery or an arterial replacement surgery that requires tissue-engineered vascular grafts. To provide insight into patterns of gene expression in endothelial cells in native or bioartificial arteries under different biomechanical conditions, this article discusses applications of whole-transcriptome sequencing in endothelial mechanobiology and vascular tissue engineering.
Collapse
|
48
|
Deng C, He Y, Feng J, Dong Z, Yao Y, Mok H, Lin M, Feng L. Extracellular matrix/stromal vascular fraction gel conditioned medium accelerates wound healing in a murine model. Wound Repair Regen 2018; 25:923-932. [PMID: 29240284 DOI: 10.1111/wrr.12602] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/15/2017] [Indexed: 12/19/2022]
Abstract
Conditioned medium (CM) is a new treatment modality in regenerative medicine and has shown a successful outcome in wound healing. We recently introduced extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel), an adipose-derived stem cell and adipose native extracellular matrix-enriched product for cytotherapy. This study aimed to evaluate the effect of CM from ECM/SVF-gel (Gel-CM) on wound healing compared with the conventional CM from adipose tissue (Adi-CM) and stem cell (SVF-CM). In vitro wound healing effect of three CMs on keratinocytes and fibroblasts was evaluated in terms of proliferation property, migratory property, and extracellular matrix production. In vivo, two full-thickness wounds were created on the back of each mice. The wounds were randomly divided to receive Gel-CM, Adi-CM, SVF-CM, and PBS injection. Histologic observations and collagen content of wound skin were made. Growth factors concentration in three CMs was further quantified. In vitro, Gel-CM promoted the proliferation and migration of keratinocytes and fibroblasts and enhanced collagen I synthesis in fibroblasts compared to Adi-CM and SVF-CM. In vivo, wound closure was faster, and dermal and epidermal regeneration was improved in the Gel-CM-treated mice compared to that in Adi-CM and SVF-CM-treated mice. Moreover, The growth factors concentration (i.e., vascular endothelial growth factor, basic fibroblast growth factor, hepatocyte growth factor, and transforming growth factor-β) in Gel-CM were significantly higher than those in Adi-CM and SVF-CM. Gel-CM generated under serum free conditions significantly enhanced wound healing effect compared to Adi-CM and SVF-CM by accelerating cell proliferation, migration, and production of ECM. This improved trophic effect may be attributed to the higher growth factors concentration in Gel-CM. Gel-CM shows potential as a novel and promising alternative to skin wound healing treatment. But limitations include the safety and immunogenicity studies of Gel-CM still remain to be clearly clarified and more data on mechanism study are needed.
Collapse
Affiliation(s)
- Chengliang Deng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China.,Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, P.R. China
| | - Yunfan He
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Ziqing Dong
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Yao Yao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Hsiaopei Mok
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Maohui Lin
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Lu Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
49
|
Abstract
Craniosynostosis is the premature fusion of the calvarial sutures that is associated with a number of physical and intellectual disabilities spanning from pediatric to adult years. Over the past two decades, techniques in molecular genetics and more recently, advances in high-throughput DNA sequencing have been used to examine the underlying pathogenesis of this disease. To date, mutations in 57 genes have been identified as causing craniosynostosis and the number of newly discovered genes is growing rapidly as a result of the advances in genomic technologies. While contributions from both genetic and environmental factors in this disease are increasingly apparent, there remains a gap in knowledge that bridges the clinical characteristics and genetic markers of craniosynostosis with their signaling pathways and mechanotransduction processes. By linking genotype to phenotype, outlining the role of cell mechanics may further uncover the specific mechanotransduction pathways underlying craniosynostosis. Here, we present a brief overview of the recent findings in craniofacial genetics and cell mechanics, discussing how this information together with animal models is advancing our understanding of craniofacial development.
Collapse
Affiliation(s)
- Zeinab Al-Rekabi
- Department of Mechanical Engineering, University of Washington, 3900 E Stevens Way NE, Seattle, WA, 98195, USA
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, 1900 9 Ave, Seattle, WA, 98101, USA
| | - Michael L. Cunningham
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, 1900 9 Ave, Seattle, WA, 98101, USA
- Department of Pediatrics, Division of Craniofacial Medicine and the, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, 3900 E Stevens Way NE, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, 3720 15 Ave NE, Seattle WA, 98105, USA
| |
Collapse
|
50
|
Shin YC, Song SJ, Hong SW, Jeong SJ, Chrzanowski W, Lee JC, Han DW. Multifaceted Biomedical Applications of Functional Graphene Nanomaterials to Coated Substrates, Patterned Arrays and Hybrid Scaffolds. NANOMATERIALS (BASEL, SWITZERLAND) 2017; 7:E369. [PMID: 29113052 PMCID: PMC5707586 DOI: 10.3390/nano7110369] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/01/2017] [Accepted: 11/01/2017] [Indexed: 12/12/2022]
Abstract
Because of recent research advances in nanoscience and nanotechnology, there has been a growing interest in functional nanomaterials for biomedical applications, such as tissue engineering scaffolds, biosensors, bioimaging agents and drug delivery carriers. Among a great number of promising candidates, graphene and its derivatives-including graphene oxide and reduced graphene oxide-have particularly attracted plenty of attention from researchers as novel nanobiomaterials. Graphene and its derivatives, two-dimensional nanomaterials, have been found to have outstanding biocompatibility and biofunctionality as well as exceptional mechanical strength, electrical conductivity and thermal stability. Therefore, tremendous studies have been devoted to employ functional graphene nanomaterials in biomedical applications. Herein, we focus on the biological potentials of functional graphene nanomaterials and summarize some of major literature concerning the multifaceted biomedical applications of functional graphene nanomaterials to coated substrates, patterned arrays and hybrid scaffolds that have been reported in recent years.
Collapse
Affiliation(s)
- Yong Cheol Shin
- Research Center for Energy Convergence Technology, Pusan National University, Busan 46241, Korea.
| | - Su-Jin Song
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan 46241, Korea.
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan 46241, Korea.
| | - Seung Jo Jeong
- GS Medical Co., Ltd., Cheongju-si, Chungcheongbuk-do 28161, Korea.
| | - Wojciech Chrzanowski
- Australian Institute for Nanoscale Science and Technology, Charles Perkins Centre, Faculty of Pharmacy, University of Sydney, Pharmacy and Bank Building A15, Sydney NSW 2006, Australia.
| | - Jae-Chang Lee
- Research Center for Industrial Chemical Biotechnology, Korea Research Institute of Chemical Technology, Ulsan 44429, Korea.
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan 46241, Korea.
| |
Collapse
|