1
|
Wongpakham T, Chunfong T, Jeamsaksiri W, Chessadangkul K, Bhanpattanakul S, Kallayanathum W, Tharasanit T, Pimpin A. Development of Pyramidal Microwells for Enhanced Cell Spheroid Formation in a Cell-on-Chip Microfluidic System for Cardiac Differentiation of Mouse Embryonic Stem Cells. Cells 2024; 13:2132. [PMID: 39768221 PMCID: PMC11674798 DOI: 10.3390/cells13242132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Three-dimensional (3D) tissue culture models provide in vivo-like conditions for studying cell physiology. This study aimed to examine the efficiency of pyramidal microwell geometries in microfluidic devices on spheroid formation, cell growth, viability, and differentiation in mouse embryonic stem cells (mESCs). The static culture using the hanging drop (HD) method served as a control. The microfluidic chips were fabricated to have varying pyramidal tip angles, including 66°, 90°, and 106°. From flow simulations, when the tip angle increased, streamline distortion decreased, resulting in more uniform flow and a lower velocity gradient near the spheroids. These findings demonstrate the significant influence of microwell geometry on fluid dynamics. The 90° microwells provide optimal conditions, including uniform flow and reduced shear stress, while maintaining the ability for waste removal, resulting in superior spheroid growth compared to the HD method and other microwell designs. From the experiments, by Day 3, spheroids in the 90° microwells reached approximately 400 µm in diameter which was significantly larger than those in the 66° microwells, 106° microwells, and HD cultures. Brachyury gene expression in the 90° microwells was four times higher than the HD method, indicating enhanced mesodermal differentiation essential for cardiac differentiation. Immunofluorescence staining confirmed cardiomyocyte differentiation. In conclusion, microwell geometry significantly influences 3D cell culture outcomes. The pyramidal microwells with a 90° tip angle proved most effective in promoting spheroid growth and cardiac differentiation of mESC differentiation, providing insights for optimizing microfluidic systems in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tepparit Wongpakham
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
| | - Thanapat Chunfong
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
| | - Wutthinan Jeamsaksiri
- Thai Microelectronics Center, National Electronics and Computer Technology Center, Chachoengsao 24000, Thailand;
| | - Kriengkai Chessadangkul
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
| | - Sudchaya Bhanpattanakul
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wirakan Kallayanathum
- Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Theerawat Tharasanit
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Alongkorn Pimpin
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
- Micro/Nano Electromechanical Integrated Device Research Unit, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
2
|
Barbato V, Genovese V, De Gregorio V, Di Nardo M, Travaglione A, De Napoli L, Fragomeni G, Zanetti EM, Adiga SK, Mondrone G, D'Hooghe T, Zheng W, Longobardi S, Catapano G, Gualtieri R, Talevi R. Dynamic in vitro culture of bovine and human ovarian tissue enhances follicle progression and health. Sci Rep 2023; 13:11773. [PMID: 37479791 PMCID: PMC10361967 DOI: 10.1038/s41598-023-37086-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/15/2023] [Indexed: 07/23/2023] Open
Abstract
In vitro ovarian cortical tissue culture, followed by culture of isolated secondary follicles, is a promising future option for production of mature oocytes. Although efforts have been made to improve the culture outcome by changing the medium composition, so far, most studies used static culture systems. Here we describe the outcome of 7 days cultures of bovine and human ovarian cortical tissue in a dynamic system using a novel perifusion bioreactor in comparison to static culture in conventional and/or gas permeable dishes. Findings show that dynamic culture significantly improves follicle quality and viability, percentage and health of secondary follicles, overall tissue health, and steroid secretion in both species. Model predictions suggest that such amelioration can be mediated by an enhanced oxygen availability and/or by fluid-mechanical shear stresses and solid compressive strains exerted on the tissue.
Collapse
Affiliation(s)
- Vincenza Barbato
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy
| | - Vincenzo Genovese
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy
- IVF Research, Education, Development S.R.L., Via Josemaria Escrivà, 68, 81100, Caserta, Italy
| | - Vincenza De Gregorio
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy
| | - Maddalena Di Nardo
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy
- Institute for Biomedical Technologies ITB, National Research Council CNR, Via Moruzzi, 1, 56124, Pisa, Italy
| | - Angela Travaglione
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy
| | - Luigi De Napoli
- Department of Mechanical, Energy and Management Engineering, University of Calabria, Via P. Bucci, 87030, Rende, CS, Italy
| | - Gionata Fragomeni
- Department of Medical and Surgical Sciences, Magna Graecia University, Viale Europa - Loc. Germaneto, 88100, Catanzaro, Italy
| | | | - Satish K Adiga
- Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576 104, India
| | - Giuseppe Mondrone
- IVF Research, Education, Development S.R.L., Via Josemaria Escrivà, 68, 81100, Caserta, Italy
| | - Thomas D'Hooghe
- Global Medical Unit Fertility, Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
- Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven (Leuven University), Gasthuisberg Campus, Herestraat 49, 3000, Leuven, Belgium
| | - Wengijng Zheng
- Global Medical Unit Fertility, Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Salvatore Longobardi
- Global Medical Unit Fertility, Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Gerardo Catapano
- Department of Mechanical, Energy and Management Engineering, University of Calabria, Via P. Bucci, 87030, Rende, CS, Italy
| | - Roberto Gualtieri
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy
| | - Riccardo Talevi
- Department of Biology, University of Naples "Federico II", Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126, Naples, Italy.
| |
Collapse
|
3
|
Dufva M. A quantitative meta-analysis comparing cell models in perfused organ on a chip with static cell cultures. Sci Rep 2023; 13:8233. [PMID: 37217582 DOI: 10.1038/s41598-023-35043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
As many consider organ on a chip for better in vitro models, it is timely to extract quantitative data from the literature to compare responses of cells under flow in chips to corresponding static incubations. Of 2828 screened articles, 464 articles described flow for cell culture and 146 contained correct controls and quantified data. Analysis of 1718 ratios between biomarkers measured in cells under flow and static cultures showed that the in all cell types, many biomarkers were unregulated by flow and only some specific biomarkers responded strongly to flow. Biomarkers in cells from the blood vessels walls, the intestine, tumours, pancreatic island, and the liver reacted most strongly to flow. Only 26 biomarkers were analysed in at least two different articles for a given cell type. Of these, the CYP3A4 activity in CaCo2 cells and PXR mRNA levels in hepatocytes were induced more than two-fold by flow. Furthermore, the reproducibility between articles was low as 52 of 95 articles did not show the same response to flow for a given biomarker. Flow showed overall very little improvements in 2D cultures but a slight improvement in 3D cultures suggesting that high density cell culture may benefit from flow. In conclusion, the gains of perfusion are relatively modest, larger gains are linked to specific biomarkers in certain cell types.
Collapse
Affiliation(s)
- Martin Dufva
- Department of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark.
| |
Collapse
|
4
|
Zare A, Salehpour A, Khoradmehr A, Bakhshalizadeh S, Najafzadeh V, Almasi-Turk S, Mahdipour M, Shirazi R, Tamadon A. Epigenetic Modification Factors and microRNAs Network Associated with Differentiation of Embryonic Stem Cells and Induced Pluripotent Stem Cells toward Cardiomyocytes: A Review. Life (Basel) 2023; 13:life13020569. [PMID: 36836926 PMCID: PMC9965891 DOI: 10.3390/life13020569] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 02/22/2023] Open
Abstract
More research is being conducted on myocardial cell treatments utilizing stem cell lines that can develop into cardiomyocytes. All of the forms of cardiac illnesses have shown to be quite amenable to treatments using embryonic (ESCs) and induced pluripotent stem cells (iPSCs). In the present study, we reviewed the differentiation of these cell types into cardiomyocytes from an epigenetic standpoint. We also provided a miRNA network that is devoted to the epigenetic commitment of stem cells toward cardiomyocyte cells and related diseases, such as congenital heart defects, comprehensively. Histone acetylation, methylation, DNA alterations, N6-methyladenosine (m6a) RNA methylation, and cardiac mitochondrial mutations are explored as potential tools for precise stem cell differentiation.
Collapse
Affiliation(s)
- Afshin Zare
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Aria Salehpour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Shabnam Bakhshalizadeh
- Reproductive Development, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Vahid Najafzadeh
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Sahar Almasi-Turk
- Department of Basic Sciences, School of Medicine, Bushehr University of Medical Sciences, Bushehr 7514633341, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran
- Correspondence: (M.M.); (R.S.); (A.T.)
| | - Reza Shirazi
- Department of Anatomy, School of Medical Sciences, Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
- Correspondence: (M.M.); (R.S.); (A.T.)
| | - Amin Tamadon
- PerciaVista R&D Co., Shiraz 7135644144, Iran
- Correspondence: (M.M.); (R.S.); (A.T.)
| |
Collapse
|
5
|
Trikalitis VD, Kroese NJJ, Kaya M, Cofiño-Fabres C, Ten Den S, Khalil ISM, Misra S, Koopman BFJM, Passier R, Schwach V, Rouwkema J. Embedded 3D printing of dilute particle suspensions into dense complex tissue fibers using shear thinning xanthan baths. Biofabrication 2022; 15. [PMID: 36347040 DOI: 10.1088/1758-5090/aca124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022]
Abstract
In order to fabricate functional organoids and microtissues, a high cell density is generally required. As such, the placement of cell suspensions in molds or microwells to allow for cell concentration by sedimentation is the current standard for the production of organoids and microtissues. Even though molds offer some level of control over the shape of the resulting microtissue, this control is limited as microtissues tend to compact towards a sphere after sedimentation of the cells. 3D bioprinting on the other hand offers complete control over the shape of the resulting structure. Even though the printing of dense cell suspensions in the ink has been reported, extruding dense cellular suspensions is challenging and generally results in high shear stresses on the cells and a poor shape fidelity of the print. As such, additional materials such as hydrogels are added in the bioink to limit shear stresses, and to improve shape fidelity and resolution. The maximum cell concentration that can be incorporated in a hydrogel-based ink before the ink's rheological properties are compromised, is significantly lower than the concentration in a tissue equivalent. Additionally, the hydrogel components often interfere with cellular self-assembly processes. To circumvent these limitations, we report a simple and inexpensive xanthan bath based embedded printing method to 3D print dense functional linear tissues using dilute particle suspensions consisting of cells, spheroids, hydrogel beads, or combinations thereof. Using this method, we demonstrated the self-organization of functional cardiac tissue fibers with a layer of epicardial cells surrounding a body of cardiomyocytes.
Collapse
Affiliation(s)
- Vasileios D Trikalitis
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Niels J J Kroese
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Mert Kaya
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands.,Surgical Robotics Laboratory, Department of Biomedical Engineering, University of Groningen and University Medical Centre Groningen, 9713AV Groningen, The Netherlands
| | - Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Simone Ten Den
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Islam S M Khalil
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands
| | - Sarthak Misra
- Surgical Robotics Laboratory, Department of Biomechanical Engineering, University of Twente, TechMed Center, MESA+ Institute, 7500AE Enschede, The Netherlands.,Surgical Robotics Laboratory, Department of Biomedical Engineering, University of Groningen and University Medical Centre Groningen, 9713AV Groningen, The Netherlands
| | - Bart F J M Koopman
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Vascularization Lab, University of Twente, Technical Medical Centre, 7500AE Enschede, The Netherlands
| |
Collapse
|
6
|
Agnoletto C, Volinia S. Mitochondria dysfunction in circulating tumor cells. Front Oncol 2022; 12:947479. [PMID: 35992829 PMCID: PMC9386562 DOI: 10.3389/fonc.2022.947479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/11/2022] [Indexed: 12/16/2022] Open
Abstract
Circulating tumor cells (CTCs) represent a subset of heterogeneous cells, which, once released from a tumor site, have the potential to give rise to metastasis in secondary sites. Recent research focused on the attempt to detect and characterize these rare cells in the circulation, and advancements in defining their molecular profile have been reported in diverse tumor species, with potential implications for clinical applications. Of note, metabolic alterations, involving mitochondria, have been implicated in the metastatic process, as key determinants in the transition of tumor cells to a mesenchymal or stemness-like phenotype, in drug resistance, and in induction of apoptosis. This review aimed to briefly analyse the most recent knowledge relative to mitochondria dysfunction in CTCs, and to envision implications of altered mitochondria in CTCs for a potential utility in clinics.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Rete Oncologica Veneta (ROV), Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Stefano Volinia
- Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Biological and Chemical Research Centre (CNBCh UW), University of Warsaw, Warsaw, Poland
- Center of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
7
|
Abdulhasan M, Ruden X, Marben T, Harris S, Ruden DM, Awonuga AO, Puscheck EE, Rappolee DA. Using Live Imaging and Fluorescence Ubiquitinated Cell Cycle Indicator Embryonic Stem Cells to Distinguish G1 Cell Cycle Delays for General Stressors like Perfluoro-Octanoic Acid and Hyperosmotic Sorbitol or G2 Cell Cycle Delay for Mutagenic Stressors like Benzo(a)pyrene. Stem Cells Dev 2022; 31:296-310. [PMID: 35678645 PMCID: PMC9232235 DOI: 10.1089/scd.2021.0330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/17/2022] [Indexed: 12/15/2022] Open
Abstract
Lowest observable adverse effects level (LOAEL) is a standard point-of-departure dose in toxicology. However, first observable adverse effects level (FOAEL) was recently reported and is used, in this study, as one criterion to detect a mutagenic stimulus in a live imager. Fluorescence ubiquitinated cell cycle indicator (FUCCI) embryonic stem cells (ESC) are green in the S-G2-M phase of the cell cycle and not green in G1-phase. Standard media change here is a mild stress that delays G1-phase and media change increases green 2.5- to 5-fold. Since stress is mild, media change rapidly increases green cell number, but higher stresses of environmental toxicants and positive control hyperosmotic stress suppress increased green after media change. Perfluoro-octanoic acid (PFOA) and diethyl phthalate (DEP) previously suppressed progression of nongreen to green cell cycle progression. Here, bisphenol A (BPA), cortisol, and positive control hyperosmotic sorbitol also suppress green fluorescence, but benzo(a)pyrene (BaP) at high doses (10 μM) increases green fluorescence throughout the 74-h exposure. Since any stress can affect many cell cycle phases, messenger RNA (mRNA) markers are best interpreted in ratios as dose-dependent mutagens increase in G2/G1 and nonmutagens increase G1/G2. After 74-h exposure, RNAseq detects G1 and G2 markers and increasing BaP doses increase G2/G1 ratios but increasing hyperosmotic sorbitol and PFOA doses increase G1/G2 marker ratios. BaP causes rapid green increase in FOAEL at 2 h of stimulus, whereas retinoic acid caused significant green fluorescence increases only late in culture. Using a live imager to establish FOAEL and G2 delay with FUCCI ESC is a new method to allow commercial and basic developmental biologists to detect drugs and environmental stimuli that are mutagenic. Furthermore, it can be used to test compounds that prevent mutations. In longitudinal studies, uniquely provided by this viable reporter and live imager protocol, follow-up can be done to test whether the preventative compound itself causes harm.
Collapse
Affiliation(s)
- Mohammed Abdulhasan
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
- Reproductive Stress 3M, Inc., Grosse Pointe Farms, Michigan, USA
| | - Ximena Ruden
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Teya Marben
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biology, College of Engineering and Science, University of Detroit Mercy, Detroit, Michigan, USA
| | - Sean Harris
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas M. Ruden
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
- Institutes for Environmental Health Science, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Awoniyi O. Awonuga
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Elizabeth E. Puscheck
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
- Reproductive Stress 3M, Inc., Grosse Pointe Farms, Michigan, USA
- Invia Fertility Clinics, Hoffman Estates, Illinois, USA
| | - Daniel A. Rappolee
- CS Mott Center for Human Growth and Development, Reproductive Endocrinology and Infertility, Department of Ob/Gyn, Wayne State University School of Medicine, Detroit, Michigan, USA
- Reproductive Stress 3M, Inc., Grosse Pointe Farms, Michigan, USA
- Program for Reproductive Sciences, Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biology, University of Windsor, Windsor, Canada
| |
Collapse
|
8
|
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
|
9
|
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: 1.3] [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
|
10
|
Endothelial mechanotransduction in cardiovascular development and regeneration: emerging approaches and animal models. CURRENT TOPICS IN MEMBRANES 2021; 87:131-151. [PMID: 34696883 PMCID: PMC9113082 DOI: 10.1016/bs.ctm.2021.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Living cells are exposed to multiple mechanical stimuli from the extracellular matrix or from surrounding cells. Mechanoreceptors are molecules that display status changes in response to mechanical stimulation, transforming physical cues into biological responses to help the cells adapt to dynamic changes of the microenvironment. Mechanical stimuli are responsible for shaping the tridimensional development and patterning of the organs in early embryonic stages. The development of the heart is one of the first morphogenetic events that occur in embryos. As the circulation is established, the vascular system is exposed to constant shear stress, which is the force created by the movement of blood. Both spatial and temporal variations in shear stress differentially modulate critical steps in heart development, such as trabeculation and compaction of the ventricular wall and the formation of the heart valves. Zebrafish embryos are small, transparent, have a short developmental period and allow for real-time visualization of a variety of fluorescently labeled proteins to recapitulate developmental dynamics. In this review, we will highlight the application of zebrafish models as a genetically tractable model for investigating cardiovascular development and regeneration. We will introduce our approaches to manipulate mechanical forces during critical stages of zebrafish heart development and in a model of vascular regeneration, as well as advances in imaging technologies to capture these processes at high resolution. Finally, we will discuss the role of molecules of the Plexin family and Piezo cation channels as major mechanosensors recently implicated in cardiac morphogenesis.
Collapse
|
11
|
Nie J, Fu J, He Y. Hydrogels: The Next Generation Body Materials for Microfluidic Chips? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003797. [PMID: 33103353 DOI: 10.1002/smll.202003797] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Indexed: 05/27/2023]
Abstract
The integration of microfluidics with biomedical research is confronted with considerable limitations due to its body materials. With high content of water, hydrogels own superior biocompatibility and degradability. Can hydrogels become another material choice for the construction of microfluidic chips, particularly biofluidics? The present review aims to systematically establish the concept of hydrogel-based microfluidic chips (HMCs) and address three main concerns: i) why choosing hydrogels? ii) how to fabricate HMCs?, and iii) in which fields to apply HMCs? It is envisioned that hydrogels may be used increasingly as substitute for traditional materials and gradually act as the body material for microfluidic chips. The modifications of conventional process are highlighted to overcome issues arising from the incompatibility between the construction methods and hydrogel materials. Specifically targeting at the "soft and wet" hydrogels, an efficient flowchart of "i) high resolution template printing; ii) damage-free demolding; iii) twice-crosslinking bonding" is proposed. Accordingly, a broader microfluidic chip concept is proposed in terms of form and function. Potential biomedical applications of HMCs are discussed. This review also highlights the challenges arising from the material replacement, as well as the future directions of the proposed concept. Finally, the authors' viewpoints and perspectives for this emerging field are discussed.
Collapse
Affiliation(s)
- Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
| |
Collapse
|
12
|
Jaberi A, Esfahani AM, Aghabaglou F, Park JS, Ndao S, Tamayol A, Yang R. Microfluidic Systems with Embedded Cell Culture Chambers for High-Throughput Biological Assays. ACS APPLIED BIO MATERIALS 2020; 3:6661-6671. [PMID: 35019392 PMCID: PMC10081828 DOI: 10.1021/acsabm.0c00439] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ability to generate chemical and mechanical gradients on chips is important for either creating biomimetic designs or enabling high-throughput assays. However, there is still a significant knowledge gap in the generation of mechanical and chemical gradients in a single device. In this study, we developed gradient-generating microfluidic circuits with integrated microchambers to allow cell culture and to introduce chemical and mechanical gradients to cultured cells. A chemical gradient is generated across the microchambers, exposing cells to a uniform concentration of drugs. The embedded microchamber also produces a mechanical gradient in the form of varied shear stresses induced upon cells among different chambers as well as within the same chamber. Cells seeded within the chambers remain viable and show a normal morphology throughout the culture time. To validate the effect of different drug concentrations and shear stresses, doxorubicin is flowed into chambers seeded with skin cancer cells at different flow rates (from 0 to 0.2 μL/min). The experimental results show that increasing doxorubicin concentration (from 0 to 30 μg/mL) within chambers not only prohibits cell growth but also induces cell death. In addition, the increased shear stress (0.005 Pa) at high flow rates poses a synergistic effect on cell viability by inducing cell damage and detachment. Moreover, the ability of the device to seed cells in a 3D microenvironment was also examined and confirmed. Collectively, the study demonstrates the potential of microchamber-embedded microfluidic gradient generators in 3D cell culture and high-throughput drug screening.
Collapse
Affiliation(s)
- Arian Jaberi
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
| | - Fariba Aghabaglou
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
| | - Jae Sung Park
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
| | - Sidy Ndao
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communications (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68516, USA
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
13
|
Curtis A, Cheng JJ, Hui EE. Cell patterning by surface tension pinning in microfluidic channels. BIOMICROFLUIDICS 2020; 14:024102. [PMID: 32161633 PMCID: PMC7058426 DOI: 10.1063/1.5140990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/14/2020] [Indexed: 05/15/2023]
Abstract
We present a simple method to pattern multiple cell populations inside a microfluidic channel. The microchannel is partially filled with a cell suspension, and the position of the liquid boundary remains pinned by surface tension. Cells then adhere only in the filled portion of the channel, producing a very sharp boundary. The process can be performed in an unmodified microfluidic channel with only a manual syringe and can be repeated multiple times to pattern cocultures or tricultures. We demonstrate the patterning method with two different mammalian cell types, 3T3 fibroblasts and NMuMG epithelial cells, and channel heights of 1.5 mm and 0.5 mm. We anticipate that this method will be useful for studies of cell-cell interactions where precise control of the fluidic microenvironment is required.
Collapse
Affiliation(s)
- Allison Curtis
- Department of Biomedical Engineering, University of California, Irvine, California 92697-2715, USA
| | | | | |
Collapse
|
14
|
Chan SW, Rizwan M, Yim EKF. Emerging Methods for Enhancing Pluripotent Stem Cell Expansion. Front Cell Dev Biol 2020; 8:70. [PMID: 32117992 PMCID: PMC7033584 DOI: 10.3389/fcell.2020.00070] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) have great potential to revolutionize the fields of tissue engineering and regenerative medicine as well as stem cell therapeutics. However, the end goal of using PSCs for therapeutic use remains distant due to limitations in current PSC production. Conventional methods for PSC expansion have limited potential to be scaled up to produce the number of cells required for the end-goal of therapeutic use due to xenogenic components, high cost or low efficiency. In this mini review, we explore novel methods and emerging technologies of improving PSC expansion: the use of the two-dimensional mechanobiological strategies of topography and stiffness and the use of three-dimensional (3D) expansion methods including encapsulation, microcarrier-based culture, and suspension culture. Additionally, we discuss the limitations of conventional PSC expansion methods as well as the challenges in implementing non-conventional methods.
Collapse
Affiliation(s)
- Sarah W. Chan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Muhammad Rizwan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
15
|
Shear stress activates ATOH8 via autocrine VEGF promoting glycolysis dependent-survival of colorectal cancer cells in the circulation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:25. [PMID: 32000836 PMCID: PMC6993408 DOI: 10.1186/s13046-020-1533-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
Background Metastasis and recurrence, wherein circulating tumour cells (CTCs) play an important role, are the leading causes of death in colorectal cancer (CRC). Metastasis-initiating CTCs manage to maintain intravascular survival under anoikis, immune attack, and importantly shear stress; however, the underlying mechanisms remain poorly understood. Methods In view of the scarcity of CTCs in the bloodstream, suspended colorectal cancer cells were flowed into the cyclic laminar shear stress (LSS) according to previous studies. Then, we detected these suspended cells with a CK8+/CD45−/DAPI+ phenotype and named them mimic circulating tumour cells (m-CTCs) for subsequent CTCs related researches. Quantitative polymerase chain reaction, western blotting, and immunofluorescence were utilised to analyse gene expression change of m-CTCs sensitive to LSS stimulation. Additionally, we examined atonal bHLH transcription factor 8 (ATOH8) expressions in CTCs among 156 CRC patients and mice by fluorescence in situ hybridisation and flow cytometry. The pro-metabolic and pro-survival functions of ATOH8 were determined by glycolysis assay, live/dead cell vitality assay, anoikis assay, and immunohistochemistry. Further, the concrete up-and-down mechanisms of m-CTC survival promotion by ATOH8 were explored. Results The m-CTCs actively responded to LSS by triggering the expression of ATOH8, a fluid mechanosensor, with executive roles in intravascular survival and metabolism plasticity. Specifically, ATOH8 was upregulated via activation of VEGFR2/AKT signalling pathway mediated by LSS induced VEGF release. ATOH8 then transcriptionally activated HK2-mediated glycolysis, thus promoting the intravascular survival of colorectal cancer cells in the circulation. Conclusions This study elucidates a novel mechanism that an LSS triggered VEGF-VEGFR2-AKT-ATOH8 signal axis mediates m-CTCs survival, thus providing a potential target for the prevention and treatment of hematogenous metastasis in CRC.
Collapse
|
16
|
Kim MH, Kino-Oka M. Bioengineering Considerations for a Nurturing Way to Enhance Scalable Expansion of Human Pluripotent Stem Cells. Biotechnol J 2020; 15:e1900314. [PMID: 31904180 DOI: 10.1002/biot.201900314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Understanding how defects in mechanotransduction affect cell-to-cell variability will add to the fundamental knowledge of human pluripotent stem cell (hPSC) culture, and may suggest new approaches for achieving a robust, reproducible, and scalable process that result in consistent product quality and yields. Here, the current state of the understanding of the fundamental mechanisms that govern the growth kinetics of hPSCs between static and dynamic cultures is reviewed, the factors causing fluctuations are identified, and culture strategies that might eliminate or minimize the occurrence of cell-to-cell variability arising from these fluctuations are discussed. The existing challenges in the development of hPSC expansion methods for enabling the transition from process development to large-scale production are addressed, a mandatory step for industrial and clinical applications of hPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
17
|
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: 1.8] [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
|
18
|
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: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Ong LJY, Ching T, Chong LH, Arora S, Li H, Hashimoto M, DasGupta R, Yuen PK, Toh YC. Self-aligning Tetris-Like (TILE) modular microfluidic platform for mimicking multi-organ interactions. LAB ON A CHIP 2019; 19:2178-2191. [PMID: 31179467 DOI: 10.1039/c9lc00160c] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Multi-organ perfusion systems offer the unique opportunity to mimic different physiological systemic interactions. However, existing multi-organ culture platforms have limited flexibility in specifying the culture conditions, device architectures, and fluidic connectivity simultaneously. Here, we report a modular microfluidic platform that addresses this limitation by enabling easy conversion of existing microfluidic devices into tissue and fluid control modules with self-aligning magnetic interconnects. This enables a 'stick-n-play' approach to assemble planar perfusion circuits that are amenable to both bioimaging-based and analytical measurements. A myriad of tissue culture and flow control TILE modules were successfully constructed with backward compatibility. Finally, we demonstrate applications in constructing recirculating multi-organ systems to emulate liver-mediated bioactivation of nutraceuticals and prodrugs to modulate their therapeutic efficacies in the context of atherosclerosis and cancer. This platform greatly facilitates the integration of existing organs-on-chip models to provide an intuitive and flexible way for users to configure different multi-organ perfusion systems.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, 117583, Singapore.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Arora S, Yim EKF, Toh YC. Environmental Specification of Pluripotent Stem Cell Derived Endothelial Cells Toward Arterial and Venous Subtypes. Front Bioeng Biotechnol 2019; 7:143. [PMID: 31259171 PMCID: PMC6587665 DOI: 10.3389/fbioe.2019.00143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are required for a multitude of cardiovascular clinical applications, such as revascularization of ischemic tissues or endothelialization of tissue engineered grafts. Patient derived primary ECs are limited in number, have donor variabilities and their in vitro phenotypes and functions can deteriorate over time. This necessitates the exploration of alternative EC sources. Although there has been a recent surge in the use of pluripotent stem cell derived endothelial cells (PSC-ECs) for various cardiovascular clinical applications, current differentiation protocols yield a heterogeneous EC population, where their specification into arterial or venous subtypes is undefined. Since arterial and venous ECs are phenotypically and functionally different, inappropriate matching of exogenous ECs to host sites can potentially affect clinical efficacy, as exemplified by venous graft mismatch when placed into an arterial environment. Therefore, there is a need to design and employ environmental cues that can effectively modulate PSC-ECs into a more homogeneous arterial or venous phenotype for better adaptation to the host environment, which will in turn contribute to better application efficacy. In this review, we will first give an overview of the developmental and functional differences between arterial and venous ECs. This provides the foundation for our subsequent discussion on the different bioengineering strategies that have been investigated to varying extent in providing biochemical and biophysical environmental cues to mature PSC-ECs into arterial or venous subtypes. The ability to efficiently leverage on a combination of biochemical and biophysical environmental cues to modulate intrinsic arterio-venous specification programs in ECs will greatly facilitate future translational applications of PSC-ECs. Since the development and maintenance of arterial and venous ECs in vivo occur in disparate physio-chemical microenvironments, it is conceivable that the application of these environmental factors in customized combinations or magnitudes can be used to selectively mature PSC-ECs into an arterial or venous subtype.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore.,Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
Regier MC, Tokar JJ, Warrick JW, Pabon L, Berthier E, Beebe DJ, Stevens KR. User-defined morphogen patterning for directing human cell fate stratification. Sci Rep 2019; 9:6433. [PMID: 31015521 PMCID: PMC6478938 DOI: 10.1038/s41598-019-42874-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Concentration gradients of biochemical stimuli such as morphogens play a critical role in directing cell fate patterning across species and throughout development but are not commonly recapitulated in vitro. While in vitro biomolecule gradients have been generated using customized microfluidic platforms, broad implementation has been limited because these platforms introduce new variables to cell culture such as externally driven flow, culture in a specialized matrix, or extended time for in situ long range diffusion. Here we introduce a method that enables preforming and then transferring user-controlled gradients to cells in standard "open" cultures. Our gradient patterning devices are modular and decoupled from the culture substrate. We find that gradient generation and transfer are predictable by finite element modeling and that device and loading parameters can be used to tune the stimulus pattern. Furthermore, we demonstrate use of these devices to spatially define morphogen signal gradients and direct peri-gastrulation fate stratification of human pluripotent stem cells. This method for extrinsic application of biochemical signal gradients can thus be used to spatially influence cellular fate decisions in a user-controlled manner.
Collapse
Affiliation(s)
- Mary C Regier
- Department of Bioengineering, University of Washington, 98195, Seattle, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jacob J Tokar
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin - Madison, 53705, Madison, USA
| | - Lil Pabon
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA
- Department of Pathology, University of Washington, 98195, Seattle, USA
| | - Erwin Berthier
- Department of Chemistry, University of Washington, 98195, Seattle, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin - Madison, 53706, Madison, USA
- Carbone Cancer Center, University of Wisconsin - Madison, 53792, Madison, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, 98195, Seattle, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 98109, Seattle, USA.
- Department of Pathology, University of Washington, 98195, Seattle, USA.
| |
Collapse
|
22
|
Jarrar YB, Jarrar Q, abed A, Abu-Shalhoob M. Effects of nonsteroidal anti-inflammatory drugs on the expression of arachidonic acid-metabolizing Cyp450 genes in mouse hearts, kidneys and livers. Prostaglandins Other Lipid Mediat 2019; 141:14-21. [DOI: 10.1016/j.prostaglandins.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/31/2019] [Accepted: 02/05/2019] [Indexed: 02/04/2023]
|
23
|
Samal P, van Blitterswijk C, Truckenmüller R, Giselbrecht S. Grow with the Flow: When Morphogenesis Meets Microfluidics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805764. [PMID: 30767289 DOI: 10.1002/adma.201805764] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Developmental biology has advanced the understanding of the intricate and dynamic processes involved in the formation of an organism from a single cell. However, many gaps remain in the knowledge of embryonic development, especially regarding tissue morphogenesis. A possible approach to mimic such phenomena uses pluripotent stem cells in in vitro morphogenetic models. Herein, these systems are summarized with emphasis on the ability to better manipulate and control cellular interfaces with either liquid or solid materials using microengineered tools, which is critical for attaining deeper insights into pattern formation and stem cell differentiation during organogenesis. The role of conventional and customized cell-culture systems in supporting important advances in the field of morphogenesis is discussed, and the fascinating role that material sciences and microengineering currently play and are expected to play in the future is highlighted. In conclusion, it is proffered that continued microfluidics innovations when applied to morphogenesis promise to provide important insights to advance many multidisciplinary fields, including regenerative medicine.
Collapse
Affiliation(s)
- Pinak Samal
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Clemens van Blitterswijk
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Roman Truckenmüller
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- Department of Complex Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
24
|
Arora S, Lam AJY, Cheung C, Yim EKF, Toh YC. Determination of critical shear stress for maturation of human pluripotent stem cell-derived endothelial cells towards an arterial subtype. Biotechnol Bioeng 2019; 116:1164-1175. [PMID: 30597522 DOI: 10.1002/bit.26910] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/29/2018] [Accepted: 12/26/2018] [Indexed: 01/07/2023]
Abstract
Human pluripotent stem cell-derived endothelial cells (hPSC-ECs) present an attractive alternative to primary EC sources for vascular grafting. However, there is a need to mature them towards either an arterial or venous subtype. A vital environmental factor involved in the arteriovenous specification of ECs during early embryonic development is fluid shear stress; therefore, there have been attempts to employ adult arterial shear stress conditions to mature hPSC-ECs. However, hPSC-ECs are naïve to fluid shear stress, and their shear responses are still not well understood. Here, we used a multiplex microfluidic platform to systematically investigate the dose-time shear responses on hPSC-EC morphology and arterial-venous phenotypes over a range of magnitudes coincidental with physiological levels of embryonic and adult vasculatures. The device comprised of six parallel cell culture chambers that were individually linked to flow-setting resistance channels, allowing us to simultaneously apply shear stress ranging from 0.4 to 15 dyne/cm 2 . We found that hPSC-ECs required up to 40 hr of shear exposure to elicit a stable phenotypic change. Cell alignment was visible at shear stress <1 dyne/cm 2 , which was independent of shear stress magnitude and duration of exposure. We discovered that the arterial markers NOTCH1 and EphrinB2 exhibited a dose-dependent increase in a similar manner beyond a threshold level of 3.8 dyne/cm 2 , whereas the venous markers COUP-TFII and EphB4 expression remained relatively constant across different magnitudes. These findings indicated that hPSC-ECs were sensitive to relatively low magnitudes of shear stress, and a critical level of ~4 dyne/cm 2 was sufficient to preferentially enhance their maturation into an arterial phenotype for future vascular tissue engineering applications.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore
| | - Adele Jing Ying Lam
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Department of Bioengineering, Imperial College London, London, UK
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technical University, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore.,Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Fabrication of a Novel Culture Dish Adapter with a Small Recess Structure for Flow Control in a Closed Environment. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9020269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cell culture medium replacement is necessary to replenish nutrients and remove waste products, and perfusion and batch media exchange methods are available. The former can establish an environment similar to that in vivo, and microfluidic devices are frequently used. However, these methods are hampered by incompatibility with commercially available circular culture dishes and the difficulty in controlling liquid flow. Here, we fabricated a culture dish adapter using polydimethylsiloxane that has a small recess structure for flow control compatible with commercially available culture dishes. We designed U-shaped and I-shaped recess structure adapters and we examined the effects of groove structure on medium flow using simulation. We found that the U-shaped and I-shaped structures allowed a uniform and uneven flow of medium, respectively. We then applied these adaptors to 293T cell culture and examined the effects of recess structures on cell proliferation. As expected, cell proliferation was similar in each area of a dish in the U-shaped structure adapter, whereas in the early flow area in the I-shaped structure adapter, it was significantly higher. In summary, we succeeded in controlling liquid flow in culture dishes with the fabricated adapter, as well as in applying the modulation of culture medium flow to control cell culture.
Collapse
|
26
|
Varma S, Voldman J. Caring for cells in microsystems: principles and practices of cell-safe device design and operation. LAB ON A CHIP 2018; 18:3333-3352. [PMID: 30324208 PMCID: PMC6254237 DOI: 10.1039/c8lc00746b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Microfluidic device designers and users continually question whether cells are 'happy' in a given microsystem or whether they are perturbed by micro-scale technologies. This issue is normally brought up by engineers building platforms, or by external reviewers (academic or commercial) comparing multiple technological approaches to a problem. Microsystems can apply combinations of biophysical and biochemical stimuli that, although essential to device operation, may damage cells in complex ways. However, assays to assess the impact of microsystems upon cells have been challenging to conduct and have led to subjective interpretation and evaluation of cell stressors, hampering development and adoption of microsystems. To this end, we introduce a framework that defines cell health, describes how device stimuli may stress cells, and contrasts approaches to measure cell stress. Importantly, we provide practical guidelines regarding device design and operation to minimize cell stress, and recommend a minimal set of quantitative assays that will enable standardization in the assessment of cell health in diverse devices. We anticipate that as microsystem designers, reviewers, and end-users enforce such guidelines, we as a community can create a set of essential principles that will further the adoption of such technologies in clinical, translational and commercial applications.
Collapse
Affiliation(s)
- Sarvesh Varma
- Department of Electrical Engineering and Computer Science
, Massachusetts Institute of Technology
,
77 Massachusetts Avenue, Room 36-824
, Cambridge
, USA
.
; Fax: +617 258 5846
; Tel: +617 253 1583
| | - Joel Voldman
- Department of Electrical Engineering and Computer Science
, Massachusetts Institute of Technology
,
77 Massachusetts Avenue, Room 36-824
, Cambridge
, USA
.
; Fax: +617 258 5846
; Tel: +617 253 1583
| |
Collapse
|
27
|
Abdolvand N, Tostoes R, Raimes W, Kumar V, Szita N, Veraitch F. Long-Term Retinal Differentiation of Human Induced Pluripotent Stem Cells in a Continuously Perfused Microfluidic Culture Device. Biotechnol J 2018; 14:e1800323. [DOI: 10.1002/biot.201800323] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/15/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Nima Abdolvand
- Department of Biochemical Engineering; University College London; Bernard Katz building London WC1E 6BT UK
| | - Rui Tostoes
- Department of Biochemical Engineering; University College London; Bernard Katz building London WC1E 6BT UK
| | - William Raimes
- Department of Biochemical Engineering; University College London; Bernard Katz building London WC1E 6BT UK
| | - Vijay Kumar
- Department of Biochemical Engineering; University College London; Bernard Katz building London WC1E 6BT UK
| | - Nicolas Szita
- Department of Biochemical Engineering; University College London; Bernard Katz building London WC1E 6BT UK
| | - Farlan Veraitch
- Department of Biochemical Engineering; University College London; Bernard Katz building London WC1E 6BT UK
| |
Collapse
|
28
|
Blastocyst-Derived Stem Cell Populations under Stress: Impact of Nutrition and Metabolism on Stem Cell Potency Loss and Miscarriage. Stem Cell Rev Rep 2018; 13:454-464. [PMID: 28425063 DOI: 10.1007/s12015-017-9734-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Data from in vitro and in vivo models suggest that malnutrition and stress trigger adaptive responses, leading to small for gestational age (SGA) blastocysts with fewer cell numbers. These stress responses are initially adaptive, but become maladaptive with increasing stress exposures. The common stress responses of the blastocyst-derived stem cells, pluripotent embryonic and multipotent placental trophoblast stem cells (ESCs and TSCs), are decreased growth and potency, and increased, imbalanced and irreversible differentiation. SGA embryos may fail to produce sufficient antiluteolytic placental hormone to maintain corpus luteum progesterone secretion that provides nutrition at the implantation site. Myriad stress inputs for the stem cells in the embryo can occur in vitro during in vitro fertilization/assisted reproductive technology (IVF/ART) or in vivo. Paradoxically, stresses that diminish stem cell growth lead to a higher level of differentiation simultaneously which further decreases ESC or TSC numbers in an attempt to functionally compensate for fewer cells. In addition, prolonged or strong stress can cause irreversible differentiation. Resultant stem cell depletion is proposed as a cause of miscarriage via a "quiet" death of an ostensibly adaptive response of stem cells instead of a reactive, violent loss of stem cells or their differentiated progenies.
Collapse
|
29
|
Wu SC, Chen CH, Wang JY, Lin YS, Chang JK, Ho ML. Hyaluronan size alters chondrogenesis of adipose-derived stem cells via the CD44/ERK/SOX-9 pathway. Acta Biomater 2018; 66:224-237. [PMID: 29128538 DOI: 10.1016/j.actbio.2017.11.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/23/2022]
Abstract
Hyaluronan (HA) is a natural linear polymer that is one of the main types of extracellular matrix during the early stage of chondrogenesis. We found that the chondrogenesis of adipose-derived stem cells (ADSCs) can be initiated and promoted by the application of HA to mimic the chondrogenic niche. The aim of this study is to investigate the optimal HA molecular weight (Mw) for chondrogenesis of ADSCs and the detailed mechanism. In this study, we investigated the relationships among HA Mw, CD44 clustering, and the extracellular signal-regulated kinase (ERK)/SOX-9 pathway during chondrogenesis of ADSCs. Human ADSCs (hADSCs) and rabbit ADSCs (rADSCs) were isolated and expanded. Chondrogenesis was induced in rADSCs by culturing cells in HA-coated wells (HA Mw: 80 kDa, 600 kDa and 2000 kDa) and evaluated by examining cell aggregation, chondrogenic gene expression (collagen type II and aggrecan) and sulfated glycosaminoglycan (sGAG) deposition in vitro. Cartilaginous tissue formation in vivo was confirmed by implanting HA/rADSCs into joint cavities. CD44 clustering, ERK phosphorylation, SOX-9 expression and SOX-9 phosphorylation in cultured hADSCs were further evaluated. Isolated and expanded rADSCs showed multilineage potential and anchorage-independent growth properties. Cell aggregation, chondrogenic gene expression, and sGAG deposition increased with increasing HA Mw in rADSCs. The 2000 kDa HA had the most pronounced chondrogenic effect on rADSCs in vitro, and implanted 2000 kDa HA/rADSCs exhibited marked cartilaginous tissue formation in vivo. CD44 clustering and cell aggregation of hADSCs were enhanced by an increase in HA Mw. In addition, higher HA Mws further enhanced CD44 clustering, ERK phosphorylation, and SOX-9 expression and phosphorylation in hADSCs. Inhibiting CD44 clustering in hADSCs reduced HA-induced chondrogenic gene expression. Inhibiting ERK phosphorylation also simultaneously attenuated HA-induced SOX-9 expression and phosphorylation and chondrogenic gene expression in hADSCs. Our results indicate that HA initiates ADSC chondrogenesis and that higher Mw HAs exhibit stronger effects, with 2000 kDa HA having the strongest effect. These effects may be mediated through increased CD44 clustering and the ERK/SOX-9 signaling pathway. STATEMENT OF SIGNIFICANCE HA-based biomaterials have been studied in stem cell-based articular cartilage tissue engineering. However, little is known about the optimal HA size for stem cell chondrogenesis and the mechanism of how HA size modulates stem cell chondrogenesis. Accordingly, we used HAs with various Mws (80-2000 kDa) as culture substrates and tested their chondrogenic effect on ADSCs. Our results demonstrated that HAs with a Mw of 2000 kDa showed the optimal effect for chondrogenesis of ADSCs. Moreover, we found that HA size can regulate ADSC chondrogenesis via the CD44/ERK/SOX-9 pathway. This finding provides new information regarding the biochemical control of chondrogenesis by HA substrates that may add value to the development of HA-based biomaterials for articular cartilage regeneration.
Collapse
Affiliation(s)
- Shun-Cheng Wu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jyun-Ya Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Mei-Ling Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
30
|
Du V, Luciani N, Richard S, Mary G, Gay C, Mazuel F, Reffay M, Menasché P, Agbulut O, Wilhelm C. A 3D magnetic tissue stretcher for remote mechanical control of embryonic stem cell differentiation. Nat Commun 2017; 8:400. [PMID: 28900152 PMCID: PMC5596024 DOI: 10.1038/s41467-017-00543-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
The ability to create a 3D tissue structure from individual cells and then to stimulate it at will is a major goal for both the biophysics and regenerative medicine communities. Here we show an integrated set of magnetic techniques that meet this challenge using embryonic stem cells (ESCs). We assessed the impact of magnetic nanoparticles internalization on ESCs viability, proliferation, pluripotency and differentiation profiles. We developed magnetic attractors capable of aggregating the cells remotely into a 3D embryoid body. This magnetic approach to embryoid body formation has no discernible impact on ESC differentiation pathways, as compared to the hanging drop method. It is also the base of the final magnetic device, composed of opposing magnetic attractors in order to form embryoid bodies in situ, then stretch them, and mechanically stimulate them at will. These stretched and cyclic purely mechanical stimulations were sufficient to drive ESCs differentiation towards the mesodermal cardiac pathway. The development of embryoid bodies that are responsive to external stimuli is of great interest in tissue engineering. Here, the authors culture embryonic stem cells with magnetic nanoparticles and show that the presence of magnetic fields could affect their aggregation and differentiation.
Collapse
Affiliation(s)
- Vicard Du
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Nathalie Luciani
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Sophie Richard
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Gaëtan Mary
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Cyprien Gay
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - François Mazuel
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Myriam Reffay
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou; Paris Cardiovascular Research Center, INSERM U970, Université Paris Descartes, Paris, 75015, France
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, 75005, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France.
| |
Collapse
|
31
|
Ong LJY, Chong LH, Jin L, Singh PK, Lee PS, Yu H, Ananthanarayanan A, Leo HL, Toh YC. A pump-free microfluidic 3D perfusion platform for the efficient differentiation of human hepatocyte-like cells. Biotechnol Bioeng 2017; 114:2360-2370. [PMID: 28542705 DOI: 10.1002/bit.26341] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 12/19/2022]
Abstract
The practical application of microfluidic liver models for in vitro drug testing is partly hampered by their reliance on human primary hepatocytes, which are limited in number and have batch-to-batch variation. Human stem cell-derived hepatocytes offer an attractive alternative cell source, although their 3D differentiation and maturation in a microfluidic platform have not yet been demonstrated. We develop a pump-free microfluidic 3D perfusion platform to achieve long-term and efficient differentiation of human liver progenitor cells into hepatocyte-like cells (HLCs). The device contains a micropillar array to immobilize cells three-dimensionally in a central cell culture compartment flanked by two side perfusion channels. Constant pump-free medium perfusion is accomplished by controlling the differential heights of horizontally orientated inlet and outlet media reservoirs. Computational fluid dynamic simulation is used to estimate the hydrostatic pressure heads required to achieve different perfusion flow rates, which are experimentally validated by micro-particle image velocimetry, as well as viability and functional assessments in a primary rat hepatocyte model. We perform on-chip differentiation of HepaRG, a human bipotent progenitor cell, and discover that 3D microperfusion greatly enhances the hepatocyte differentiation efficiency over static 2D and 3D cultures. However, HepaRG progenitor cells are highly sensitive to the time-point at which microperfusion is applied. Isolated HepaRG cells that are primed as static 3D spheroids before being subjected to microperfusion yield a significantly higher proportion of HLCs (92%) than direct microperfusion of isolated HepaRG cells (62%). This platform potentially offers a simple and efficient means to develop highly functional microfluidic liver models incorporating human stem cell-derived HLCs. Biotechnol. Bioeng. 2017;114: 2360-2370. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore, 117583
| | - Lor Huai Chong
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore, 117583
| | - Lin Jin
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore, 117583
| | - Pawan Kumar Singh
- Department of Mechanical Engineering, National University of Singapore, Singapore
| | - Poh Seng Lee
- Department of Mechanical Engineering, National University of Singapore, Singapore
| | - Hanry Yu
- Department of Physiology, National University of Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, Singapore.,Mechanobiology Institute, National University of Singapore, Singapore
| | | | - Hwa Liang Leo
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore, 117583
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore, 117583.,Singapore Institute for Neurotechnology, Singapore.,NUS Tissue Engineering Programme, National University of Singapore, Singapore
| |
Collapse
|
32
|
A method for high-throughput functional imaging of single cells within heterogeneous cell preparations. Sci Rep 2016; 6:39319. [PMID: 27982116 PMCID: PMC5159830 DOI: 10.1038/srep39319] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/17/2016] [Indexed: 01/25/2023] Open
Abstract
Functional characterization of individual cells within heterogeneous tissue preparations is challenging. Here, we report the development of a versatile imaging method that assesses single cell responses of various endpoints in real time, while identifying the individual cell types. Endpoints that can be measured include (but are not limited to) ionic flux (calcium, sodium, potassium and hydrogen), metabolic responsiveness (NAD(P)H, mitochondrial membrane potential), and signal transduction (H2O2 and cAMP). Subsequent to fluorescent imaging, identification of cell types using immunohistochemistry allows for mapping of cell type to their respective functional real time responses. To validate the utility of this method, NAD(P)H responses to glucose of islet alpha versus beta cells generated from dispersed pancreatic islets, followed by the construction of frequency distributions characterizing the variability in the magnitude of each individual cell responses were compared. As expected, no overlap between the glucose response frequency distributions for beta cells versus alpha cells was observed, thereby establishing both the high degree of fidelity and low rate of both false-negatives and false-positives in this approach. This novel method has the ability not only to resolve single cell level functional differences between cell types, but also to characterize functional heterogeneity within a given cell type.
Collapse
|
33
|
Yang Y, Jiang Z, Bolnick A, Dai J, Puscheck EE, Rappolee DA. Departure from optimal O 2 level for mouse trophoblast stem cell proliferation and potency leads to most rapid AMPK activation. J Reprod Dev 2016; 63:87-94. [PMID: 27867161 PMCID: PMC5320434 DOI: 10.1262/jrd.2016-110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Previous studies showed that cultured mouse trophoblast stem cells (mTSCs) have the most rapid proliferation, normal maintenance of stemness/potency, the
least spontaneous differentiation, and the lowest level of stress-activated protein kinase (SAPK) when incubated at 2% O2 rather than at the
traditional 20% O2 or hypoxic (0.5% and 0% O2) conditions. Switching from 2% O2 induced fast SAPK responses. Here we tested the
dose response of AMP-activated protein kinase (AMPK) in its active form (pAMPK Thr172P) at O2 levels from 20–0%, and also tested whether pAMPK levels
show similar rapid changes when mTSC cultures were switched from the optimal 2% O2 to other O2 conditions. There was a delayed increase in
pAMPK levels ~6–8 h after switching conditions from 20% to 2%, 0.5%, or 0% O2. Altering O2 conditions from 2% to either 20%, 0.5%, or 0%
led to rapid increase in pAMPK levels within 1 h, similar to the previously reported SAPK response in mTSC cells removed from 2% O2. Twelve hours of
0.5% O2 exposure led to cell program changes in terms of potency loss and suppressed biosynthesis, as indicated by levels of phosphorylated inactive
acetyl CoA carboxylase (pACC). Phosphorylation of ACC was inhibited by the AMPK inhibitor Compound C. However, unlike other stressors, AMPK does not mediate
hypoxia-induced potency loss in mTSCs. These results suggest an important aspect of stem cell biology, which demands rapid stress enzyme activation to cope with
sudden changes in external environment, e.g., from least stressful (2% O2) to more stressful conditions.
Collapse
Affiliation(s)
- Yu Yang
- Ob/Gyn, Wayne State University Medical School, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
34
|
Przybyla L, Muncie JM, Weaver VM. Mechanical Control of Epithelial-to-Mesenchymal Transitions in Development and Cancer. Annu Rev Cell Dev Biol 2016; 32:527-554. [DOI: 10.1146/annurev-cellbio-111315-125150] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Laralynne Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
| | - Jonathon M. Muncie
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
- Joint Graduate Group in Bioengineering (University of California, San Francisco, and University of California, Berkeley), San Francisco, California 94143
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
- Departments of Anatomy, Bioengineering, and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143
| |
Collapse
|
35
|
Huang C, Melerzanov A, Du Y. Engineering Embryonic Stem Cell Microenvironments for Tailored Cellular Differentiation. J Nanotechnol Eng Med 2016. [DOI: 10.1115/1.4033193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The rapid progress of embryonic stem cell (ESCs) research offers great promise for drug discovery, tissue engineering, and regenerative medicine. However, a major limitation in translation of ESCs technology to pharmaceutical and clinical applications is how to induce their differentiation into tailored lineage commitment with satisfactory efficiency. Many studies indicate that this lineage commitment is precisely controlled by the ESC microenvironment in vivo. Engineering and biomaterial-based approaches to recreate a biomimetic cellular microenvironment provide valuable strategies for directing ESCs differentiation to specific lineages in vitro. In this review, we summarize and examine the recent advances in application of engineering and biomaterial-based approaches to control ESC differentiation. We focus on physical strategies (e.g., geometrical constraint, mechanical stimulation, extracellular matrix (ECM) stiffness, and topography) and biochemical approaches (e.g., genetic engineering, soluble bioactive factors, coculture, and synthetic small molecules), and highlight the three-dimensional (3D) hydrogel-based microenvironment for directed ESC differentiation. Finally, future perspectives in ESCs engineering are provided for the subsequent advancement of this promising research direction.
Collapse
Affiliation(s)
- Chenyu Huang
- Department of Plastic, Reconstructive and Aesthetic Surgery, Beijing Tsinghua Changgung Hospital, Medical Center, Tsinghua University, Beijing 100084, China
- Department of Plastic Surgery, Meitan General Hospital, Beijing 100028, China e-mail:
| | - Alexander Melerzanov
- Cellular and Molecular Technologies Laboratory, MIPT, Dolgoprudny 141701, Russia
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China e-mail:
| |
Collapse
|
36
|
Rodriguez G, Pieralisi I, Anderlei T, Ducci A, Micheletti M. Appraisal of fluid flow in a shaken bioreactor with conical bottom at different operating conditions. Chem Eng Res Des 2016. [DOI: 10.1016/j.cherd.2015.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Youssef K, Jarenwattananon NN, Archer BJ, Mack J, Iruela-Arispe ML, Bouchard LS. 4-D Flow Control in Porous Scaffolds: Toward a Next Generation of Bioreactors. IEEE Trans Biomed Eng 2016; 64:61-69. [PMID: 26955013 DOI: 10.1109/tbme.2016.2537266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tissue engineering (TE) approaches that involve seeding cells into predetermined tissue scaffolds ignore the complex environment where the material properties are spatially inhomogeneous and evolve over time. We present a new approach for controlling mechanical forces inside bioreactors, which enables spatiotemporal control of flow fields in real time. Our adaptive approach offers the flexibility of dialing-in arbitrary shear stress distributions and adjusting flow field patterns in a scaffold over time in response to cell growth without needing to alter scaffold structure. This is achieved with a multi-inlet bioreactor and a control algorithm with learning capabilities to dynamically solve the inverse problem of computing the inlet pressure distribution required over the multiple inlets to obtain a target flow field. The new method constitutes a new platform for studies of cellular responses to mechanical forces in complex environments and opens potentially transformative possibilities for TE.
Collapse
|
38
|
Ghaemi RV, Vahidi B, Sabour MH, Haghighipour N, Alihemmati Z. Fluid-Structure Interactions Analysis of Shear-Induced Modulation of a Mesenchymal Stem Cell: An Image-Based Study. Artif Organs 2016; 40:278-287. [PMID: 26333040 DOI: 10.1111/aor.12547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although effects of biochemical modulation of stem cells have been widely investigated, only recent advances have been made in the identification of mechanical conditioning on cell signaling pathways. Experimental investigations quantifying the micromechanical environment of mesenchymal stem cells (MSCs) are challenging while computational approaches can predict their behavior due to in vitro stimulations. This study introduces a 3D cell-specific finite element model simulating large deformations of MSCs. Here emphasizing cell mechanical modulation which represents the most challenging multiphysics phenomena in sub-cellular level, we focused on an approach attempting to elicit unique responses of a cell under fluid flow. Fluorescent staining of MSCs was performed in order to visualize the MSC morphology and develop a geometrically accurate model of it based on a confocal 3D image. We developed a 3D model of a cell fixed in a microchannel under fluid flow and then solved the numerical model by fluid-structure interactions method. By imposing flow characteristics representative of vigorous in vitro conditions, the model predicts that the employed external flow induces significant localized effective stress in the nucleo-cytoplasmic interface and average cell deformation of about 40%. Moreover, it can be concluded that a lower strain level is made in the cell by the oscillatory flow as compared with steady flow, while same ranges of effective stress are recorded inside the cell in both conditions. The deeper understanding provided by this study is beneficial for better design of single cell in vitro studies.
Collapse
Affiliation(s)
- Roza Vaez Ghaemi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
- Department of Chemical and Biological Engineering, Faculty of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Bahman Vahidi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mohammad Hossein Sabour
- Department of Aerospace Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | | | - Zakieh Alihemmati
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
39
|
Abstract
Thorough understanding of the effects of shear stress on stem cells is critical for the rationale design of large-scale production of cell-based therapies. This is of growing importance as emerging tissue engineering and regenerative medicine applications drive the need for clinically relevant numbers of both pluripotent stem cells (PSCs) and cells derived from PSCs. Here, we describe the use of a custom parallel plate bioreactor system to impose fluid shear stress on a layer of PSCs adhered to protein-coated glass slides. This system can be useful both for basic science studies in mechanotransduction and as a surrogate model for bioreactors used in large-scale production.
Collapse
Affiliation(s)
- Russell P Wolfe
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA
| | - Julia B Guidry
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA
| | - Stephanie L Messina
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA
| | - Tabassum Ahsan
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA.
| |
Collapse
|
40
|
Shemesh J, Jalilian I, Shi A, Heng Yeoh G, Knothe Tate ML, Ebrahimi Warkiani M. Flow-induced stress on adherent cells in microfluidic devices. LAB ON A CHIP 2015; 15:4114-27. [PMID: 26334370 DOI: 10.1039/c5lc00633c] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Transduction of mechanical forces and chemical signals affect every cell in the human body. Fluid flow in systems such as the lymphatic or circulatory systems modulates not only cell morphology, but also gene expression patterns, extracellular matrix protein secretion and cell-cell and cell-matrix adhesions. Similar to the role of mechanical forces in adaptation of tissues, shear fluid flow orchestrates collective behaviours of adherent cells found at the interface between tissues and their fluidic environments. These behaviours range from alignment of endothelial cells in the direction of flow to stem cell lineage commitment. Therefore, it is important to characterize quantitatively fluid interface-dependent cell activity. Common macro-scale techniques, such as the parallel plate flow chamber and vertical-step flow methods that apply fluid-induced stress on adherent cells, offer standardization, repeatability and ease of operation. However, in order to achieve improved control over a cell's microenvironment, additional microscale-based techniques are needed. The use of microfluidics for this has been recognized, but its true potential has emerged only recently with the advent of hybrid systems, offering increased throughput, multicellular interactions, substrate functionalization on 3D geometries, and simultaneous control over chemical and mechanical stimulation. In this review, we discuss recent advances in microfluidic flow systems for adherent cells and elaborate on their suitability to mimic physiologic micromechanical environments subjected to fluid flow. We describe device design considerations in light of ongoing discoveries in mechanobiology and point to future trends of this promising technology.
Collapse
Affiliation(s)
- Jonathan Shemesh
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | | | | | |
Collapse
|
41
|
Recent advances and future applications of microfluidic live-cell microarrays. Biotechnol Adv 2015; 33:948-61. [DOI: 10.1016/j.biotechadv.2015.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/16/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022]
|
42
|
Abstract
PURPOSE OF REVIEW Blood flow is intimately linked with cardiovascular development, repair and dysfunction. The current review will build on the fluid mechanical principle underlying haemodynamic shear forces, mechanotransduction and metabolic effects. RECENT FINDINGS Pulsatile flow produces both time (∂τ/∂t) and spatial-varying shear stress (∂τ/∂x) to modulate vascular oxidative stress and inflammatory response with pathophysiological significance to atherosclerosis. The characteristics of haemodynamic shear forces, namely, steady laminar (∂τ/∂t = 0), pulsatile shear stress (PSS: unidirectional forward flow) and oscillatory shear stress (bidirectional with a near net 0 forward flow), modulate mechano-signal transduction to influence metabolic effects on vascular endothelial function. Atheroprotective PSS promotes antioxidant, anti-inflammatory and antithrombotic responses, whereas atherogenic oscillatory shear stress induces nicotinamide adenine dinucleotide phosphate oxidase-JNK signalling to increase mitochondrial superoxide production, protein degradation of manganese superoxide dismutase and post-translational protein modifications of LDL particles in the disturbed flow-exposed regions of vasculature. In the era of tissue regeneration, shear stress has been implicated in reactivation of developmental genes, namely, Wnt and Notch signalling, for vascular development and repair. SUMMARY Blood flow imparts a dynamic continuum from vascular development to repair. Augmentation of PSS confers atheroprotection and reactivation of developmental signalling pathways for regeneration.
Collapse
Affiliation(s)
- Juhyun Lee
- Department of Bioengineering, Department of Medicine, all at the University of California, Los Angeles, Los Angeles, California
| | - René R. Sevag Packard
- Department of Molecular, Cellular and Integrative Physiology, Department of Medicine, all at the University of California, Los Angeles, Los Angeles, California
- Division of Cardiology, Department of Medicine, all at the University of California, Los Angeles, Los Angeles, California
| | - Tzung K. Hsiai
- Department of Bioengineering, Department of Medicine, all at the University of California, Los Angeles, Los Angeles, California
- Department of Molecular, Cellular and Integrative Physiology, Department of Medicine, all at the University of California, Los Angeles, Los Angeles, California
- Division of Cardiology, Department of Medicine, all at the University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
43
|
Controlled electromechanical cell stimulation on-a-chip. Sci Rep 2015; 5:11800. [PMID: 26135970 PMCID: PMC4488866 DOI: 10.1038/srep11800] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/28/2015] [Indexed: 12/15/2022] Open
Abstract
Stem cell research has yielded promising advances in regenerative medicine, but standard assays generally lack the ability to combine different cell stimulations with rapid sample processing and precise fluid control. In this work, we describe the design and fabrication of a micro-scale cell stimulator capable of simultaneously providing mechanical, electrical, and biochemical stimulation, and subsequently extracting detailed morphological and gene-expression analysis on the cellular response. This micro-device offers the opportunity to overcome previous limitations and recreate critical elements of the in vivo microenvironment in order to investigate cellular responses to three different stimulations. The platform was validated in experiments using human bone marrow mesenchymal stem cells. These experiments demonstrated the ability for inducing changes in cell morphology, cytoskeletal fiber orientation and changes in gene expression under physiological stimuli. This novel bioengineering approach can be readily applied to various studies, especially in the fields of stem cell biology and regenerative medicine.
Collapse
|
44
|
Jabart E, Rangarajan S, Lieu C, Hack J, Conboy I, Sohn LL. A Microfluidic Method for the Selection of Undifferentiated Human Embryonic Stem Cells and in Situ Analysis. MICROFLUIDICS AND NANOFLUIDICS 2015; 18:955-966. [PMID: 33688311 PMCID: PMC7939131 DOI: 10.1007/s10404-014-1485-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Conventional cell-sorting methods such as fluorescence-activated cell sorting (FACS) or magnetic-activated cell sorting (MACS) can suffer from certain shortcomings such as lengthy sample preparation time, cell modification through antibody labeling, and cell damage due to exposure to high shear forces or to attachment of superparamagnetic Microbeads. In light of these drawbacks, we have recently developed a label-free, microfluidic platform that can not only select cells with minimal sample preparation but also enable analysis of cells in situ. We demonstrate the utility of our platform by successfully isolating undifferentiated human embryonic stem cells (hESCs) from a heterogeneous population based on the undifferentiated stem-cell marker SSEA-4. Importantly, we show that, in contrast to MACS or FACS, cells isolated by our method have very high viability (~90%). Overall, our platform technology could likely be applied to other cell types beyond hESCs and to a variety of heterogeneous cell populations in order to select and analyze cells of interest.
Collapse
Affiliation(s)
- E. Jabart
- Dept. of Bioengineering, University of California, Berkeley 94720, USA
| | - S. Rangarajan
- Dept. of Bioengineering, University of California, Berkeley 94720, USA
| | - C. Lieu
- School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - J. Hack
- Dept. of Mechanical Engineering, University of California, Berkeley 94720, USA
| | - I. Conboy
- Dept. of Bioengineering, University of California, Berkeley 94720, USA
| | - L. L. Sohn
- Dept. of Mechanical Engineering, University of California, Berkeley 94720, USA
- Author to whom correspondence should be addressed: , 510-642-5434
| |
Collapse
|
45
|
Varma S, Voldman J. A cell-based sensor of fluid shear stress for microfluidics. LAB ON A CHIP 2015; 15:1563-73. [PMID: 25648195 PMCID: PMC4443851 DOI: 10.1039/c4lc01369g] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Microsystems designed for cell-based studies or applications inherently require fluid handling. Flows within such systems inevitably generate fluid shear stress (FSS) that may adversely affect cell health. Simple assays of cell viability, morphology or growth are typically reported to indicate any gross disturbances to cell physiology. However, no straightforward metric exists to specifically evaluate physiological implications of FSS within microfluidic devices, or among competing microfluidic technologies. This paper presents the first genetically encoded cell sensors that fluoresce in a quantitative fashion upon FSS pathway activation. We picked a widely used cell line (NIH3T3s) and created a transcriptional cell-sensor where fluorescence turns on when transcription of a relevant FSS-induced protein is initiated. Specifically, we chose Early Growth Factor-1 (a mechanosensitive protein) upregulation as the node for FSS detection. We verified our sensor pathway specificity and functionality by noting induced fluorescence in response to chemical induction of the FSS pathway, seen both through microscopy and flow cytometry. Importantly, we found our cell sensors to be inducible by a range of FSS intensities and durations, with a limit of detection of 2 dynes cm(-2) when applied for 30 minutes. Additionally, our cell-sensors proved their versatility by showing induction sensitivity when made to flow through an inertial microfluidic device environment with typical flow conditions. We anticipate these cell sensors to have wide application in the microsystems community, allowing the device designer to engineer systems with acceptable FSS, and enabling the end-user to evaluate the impact of FSS upon their assay of interest.
Collapse
Affiliation(s)
- Sarvesh Varma
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Room 36-824, Cambridge, USA.
| | | |
Collapse
|
46
|
Chen H, Sun J, Wolvetang E, Cooper-White J. High-throughput, deterministic single cell trapping and long-term clonal cell culture in microfluidic devices. LAB ON A CHIP 2015; 15:1072-83. [PMID: 25519528 DOI: 10.1039/c4lc01176g] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
We report the design and validation of a two-layered microfluidic device platform for single cell capture, culture and clonal expansion. Under manual injection of a cell suspension, deterministic trapping of hundreds to thousands of single cells (adherent and non-adherent) in a high throughput manner and at high trapping efficiency was achieved simply through the incorporation of a U-shaped hydrodynamic trap into the downstream wall of each micro-well. Post single cell trapping, we confirmed that these modified micro-wells permit the attachment, spreading and proliferation of the trapped single cells for multiple generations over extended periods of time (>7 days) under media perfusion. Due to its a) low cost, b) simplicity in fabrication and operation, c) high trapping efficiency, d) reliable and repeatable trapping mechanism, e) cell size selection and f) capability to provide perfused long-term culture and continuous time-lapse imaging, the microfluidic device developed and validated in this study is seen to have significant potential application in high-throughput single cell quality assessment and clonal analysis.
Collapse
Affiliation(s)
- Huaying Chen
- Tissue Engineering and Microfluidics Laboratory, Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | | | | | | |
Collapse
|
47
|
Abstract
Anchorage-dependent cells are of great interest for various biotechnological applications. (i) They represent a formidable production means of viruses for vaccination purposes at very large scales (in 1000-6000 l reactors) using microcarriers, and in the last decade many more novel viral vaccines have been developed using this production technology. (ii) With the advent of stem cells and their use/potential use in clinics for cell therapy and regenerative medicine purposes, the development of novel culture devices and technologies for adherent cells has accelerated greatly with a view to the large-scale expansion of these cells. Presently, the really scalable systems--microcarrier/microcarrier-clump cultures using stirred-tank reactors--for the expansion of stem cells are still in their infancy. Only laboratory scale reactors of maximally 2.5 l working volume have been evaluated because thorough knowledge and basic understanding of critical issues with respect to cell expansion while retaining pluripotency and differentiation potential, and the impact of the culture environment on stem cell fate, etc., are still lacking and require further studies. This article gives an overview on critical issues common to all cell culture systems for adherent cells as well as specifics for different types of stem cells in view of small- and large-scale cell expansion and production processes.
Collapse
|
48
|
Geuss LR, Wu DC, Ramamoorthy D, Alford CD, Suggs LJ. Paramagnetic beads and magnetically mediated strain enhance cardiomyogenesis in mouse embryoid bodies. PLoS One 2014; 9:e113982. [PMID: 25501004 PMCID: PMC4264692 DOI: 10.1371/journal.pone.0113982] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/25/2014] [Indexed: 01/16/2023] Open
Abstract
Mechanical forces play an important role in proper embryologic development, and similarly such forces can directly impact pluripotency and differentiation of mouse embryonic stem cells (mESC) in vitro. In addition, manipulation of the embryoid body (EB) microenvironment, such as by incorporation of microspheres or microparticles, can similarly influence fate determination. In this study, we developed a mechanical stimulation regimen using permanent neodymium magnets to magnetically attract cells within an EB. Arginine-Glycine-Aspartic Acid (RGD)-conjugated paramagnetic beads were incorporated into the interior of the EBs during aggregation, allowing us to exert force on individual cells using short-term magnetization. EBs were stimulated for one hour at different magnetic field strengths, subsequently exerting a range of force intensity on the cells at different stages of early EB development. Our results demonstrated that following exposure to a 0.2 Tesla magnetic field, ESCs respond to magnetically mediated strain by activating Protein Kinase A (PKA) and increasing phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) expression. The timing of stimulation can also be tailored to guide ESC differentiation: the combination of bone morphogenetic protein 4 (BMP4) supplementation with one hour of magnetic attraction on Day 3 enhances cardiomyogenesis by increasing contractile activity and the percentage of sarcomeric α-actin-expressing cells compared to control samples with BMP4 alone. Interestingly, we also observed that the beads alone had some impact on differentiation by increasingly slightly, albeit not significantly, the percentage of cardiomyocytes. Together these results suggest that magnetically mediated strain can be used to enhance the percentage of mouse ESC-derived cardiomyocytes over current differentiation protocols.
Collapse
Affiliation(s)
- Laura R. Geuss
- The University of Texas at Austin, Institute of Cell and Molecular Biology, Austin, Texas, United States of America
| | - Douglas C. Wu
- The University of Texas at Austin, Institute of Cell and Molecular Biology, Austin, Texas, United States of America
| | - Divya Ramamoorthy
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States of America
| | - Corinne D. Alford
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States of America
| | - Laura J. Suggs
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
49
|
Li R, Beebe T, Jen N, Yu F, Takabe W, Harrison M, Cao H, Lee J, Yang H, Han P, Wang K, Shimizu H, Chen J, Lien CL, Chi NC, Hsiai TK. Shear stress-activated Wnt-angiopoietin-2 signaling recapitulates vascular repair in zebrafish embryos. Arterioscler Thromb Vasc Biol 2014; 34:2268-75. [PMID: 25147335 DOI: 10.1161/atvbaha.114.303345] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Fluid shear stress intimately regulates vasculogenesis and endothelial homeostasis. The canonical Wnt/β-catenin signaling pathways play an important role in differentiation and proliferation. In this study, we investigated whether shear stress activated angiopoietin-2 (Ang-2) via the canonical Wnt signaling pathway with an implication in vascular endothelial repair. APPROACH AND RESULTS Oscillatory shear stress upregulated both TOPflash Wnt reporter activities and the expression of Ang-2 mRNA and protein in human aortic endothelial cells accompanied by an increase in nuclear β-catenin intensity. Oscillatory shear stress-induced Ang-2 and Axin-2 mRNA expression was downregulated in the presence of a Wnt inhibitor, IWR-1, but was upregulated in the presence of a Wnt agonist, LiCl. Ang-2 expression was further downregulated in response to a Wnt signaling inhibitor, DKK-1, but was upregulated by Wnt agonist Wnt3a. Both DKK-1 and Ang-2 siRNA inhibited endothelial cell migration and tube formation, which were rescued by human recombinant Ang-2. Both Ang-2 and Axin-2 mRNA downregulation was recapitulated in the heat-shock-inducible transgenic Tg(hsp70l:dkk1-GFP) zebrafish embryos at 72 hours post fertilization. Ang-2 morpholino injection of Tg (kdrl:GFP) fish impaired subintestinal vessel formation at 72 hours post fertilization, which was rescued by zebrafish Ang-2 mRNA coinjection. Inhibition of Wnt signaling with IWR-1 also downregulated Ang-2 and Axin-2 expression and impaired vascular repair after tail amputation, which was rescued by zebrafish Ang-2 mRNA injection. CONCLUSIONS Shear stress activated Ang-2 via canonical Wnt signaling in vascular endothelial cells, and Wnt-Ang-2 signaling is recapitulated in zebrafish embryos with a translational implication in vascular development and repair.
Collapse
Affiliation(s)
- Rongsong Li
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Tyler Beebe
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Nelson Jen
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Fei Yu
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Wakako Takabe
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Michael Harrison
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Hung Cao
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Juhyun Lee
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Hongbo Yang
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Peidong Han
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Kevin Wang
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Hirohito Shimizu
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Jaunian Chen
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Ching-Ling Lien
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Neil C Chi
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla
| | - Tzung K Hsiai
- From the Department of Medicine, School of Medicine (R.L., T.K.H.), Department of Bioengineering (T.B., N.J., F.Y., W.T., H.C., J.L., T.K.H.), and Department of Molecular, Cell, and Developmental Biology (K.W., H.S., J.C.), University of California, Los Angeles; Department of Surgery, Children's Hospital Los Angeles, CA (M.H., C.-L.L.); and Division of Cardiology, Department of Medicine, School of Medicine (H.Y., P.H., N.C.C.) and Institute of Genomic Medicine (N.C.C.), University of California, San Diego, La Jolla.
| |
Collapse
|
50
|
Morley WA, Seneff S. Diminished brain resilience syndrome: A modern day neurological pathology of increased susceptibility to mild brain trauma, concussion, and downstream neurodegeneration. Surg Neurol Int 2014; 5:97. [PMID: 25024897 PMCID: PMC4093745 DOI: 10.4103/2152-7806.134731] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/31/2014] [Indexed: 12/11/2022] Open
Abstract
The number of sports-related concussions has been steadily rising in recent years. Diminished brain resilience syndrome is a term coined by the lead author to describe a particular physiological state of nutrient functional deficiency and disrupted homeostatic mechanisms leading to increased susceptibility to previously considered innocuous concussion. We discuss how modern day environmental toxicant exposure, along with major changes in our food supply and lifestyle practices, profoundly reduce the bioavailability of neuro-critical nutrients such that the normal processes of homeostatic balance and resilience are no longer functional. Their diminished capacity triggers physiological and biochemical 'work around' processes that result in undesirable downstream consequences. Exposure to certain environmental chemicals, particularly glyphosate, the active ingredient in the herbicide, Roundup(®), may disrupt the body's innate switching mechanism, which normally turns off the immune response to brain injury once danger has been removed. Deficiencies in serotonin, due to disruption of the shikimate pathway, may lead to impaired melatonin supply, which reduces the resiliency of the brain through reduced antioxidant capacity and alterations in the cerebrospinal fluid, reducing critical protective buffering mechanisms in impact trauma. Depletion of certain rare minerals, overuse of sunscreen and/or overprotection from sun exposure, as well as overindulgence in heavily processed, nutrient deficient foods, further compromise the brain's resilience. Modifications to lifestyle practices, if widely implemented, could significantly reduce this trend of neurological damage.
Collapse
Affiliation(s)
| | - Stephanie Seneff
- Spoken Language Systems Group, Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge MA 02139, USA
| |
Collapse
|