1
|
Yamatoya K, Nagai Y, Teramoto N, Kang W, Miyado K, Nakata K, Yagi T, Miyamoto Y. Dimethyl Sulfoxide-Free Cryopreservation of Differentiated Human Neuronal Cells. Biopreserv Biobank 2023; 21:631-634. [PMID: 36827090 DOI: 10.1089/bio.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
In recent years, cells provided by cell banks and medical facilities have been used for cell therapy, regenerative therapy, and fundamental research. Cryopreservation is an effective means of maintaining stable cell quality over a long period of time. The slow freezing method is most suitable for processing many human cells isolated simultaneously from organs and tissues, but it is necessary to develop a freezing solution for this method. In this study, we report the successful development of a dimethyl sulfoxide (DMSO)-free freezing medium for differentiated neuronal cells. Neuronal differentiation results in the differentiation of undifferentiated SK-N-SH cells into neuronal cells. A basic freezing medium (BFM) was prepared using Dulbecco's modified Eagle's medium, 1 M maltose, and 1% sericin as the essential ingredients, supplemented with 5%-40% propylene glycol (PG). Each BFM supplemented with 5%-40% PG was evaluated in undifferentiated cells. After thawing, BFM supplemented with 10% and 20% PG were 83% and 88% viable, respectively. There was no significant difference between the 10% and 20% PG groups. However, a significant difference was observed when the concentration of PG in the BFM decreased by 5% (5% PG vs. 10% PG; p = 0.0026). Each DMSO-free BFM was evaluated using differentiated neuronal cells. There was no significant difference between the 10% PG BFM and stem-CB-free groups. Viability was significantly different in the 10% glycerol BFM (4.8%) and 10% PG BFM (45%) (p = 0.028). The differentiated cells with 10% PG BFM showed higher adherence to culture dishes than those with 10% glycerol BFM. These results show that BFM containing PG was effective in differentiating neuronal cells. DMSO affects the central nervous system at low concentrations. This report indicates that DMSO is unsuitable for neuronal cells with multipotent differentiation potential. Therefore, it is essential for cell banking and transplantation medicine services to select appropriate cell freezing media.
Collapse
Affiliation(s)
- Kenji Yamatoya
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
- Laboratory of Genomic Function Engineering, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, Japan
| | - Yuya Nagai
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Naozumi Teramoto
- Department of Applied Chemistry, Faculty of Engineering, Chiba Institute of Technology, Narashino, Japan
| | - Woojin Kang
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya-ku, Japan
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya-ku, Japan
| | - Kazuya Nakata
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
- Division of Sciences for Biological System, Institute of Agriculture, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Tohru Yagi
- Department of Mechanical Engineering, Tokyo Institute of Technology, Meguro-ku, Japan
| | - Yoshitaka Miyamoto
- Department of Applied Chemistry, Faculty of Engineering, Chiba Institute of Technology, Narashino, Japan
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya-ku, Japan
- Department of Mechanical Engineering, Tokyo Institute of Technology, Meguro-ku, Japan
| |
Collapse
|
2
|
Jiang P, Li Q, Liu B, Liang W. Effect of cryoprotectant-induced intracellular ice formation and crystallinity on bactria during cryopreservation. Cryobiology 2023; 113:104786. [PMID: 37863380 DOI: 10.1016/j.cryobiol.2023.104786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Cryopreservation is widely used for the long-term storage of bacteria. Glycerol is one of the traditional cryoprotectants used widely to prevent cryoinjury during the cryopreservation of bacteria,although it may be toxic to the cells. To overcome these issues, synthetic antifreeze polymers are also used as cryoprotectants to inhibit ice formation. In the study, we compared the performance of various antifreeze synthetic polymers including poly(vinyl alcohol) (PVA), poly(vinylpyrrolidone), poly(ethylene glycol), and dextran with glycerol, among which PVA performed best on decreasing the ice growth rate.The impacts of glycerol, trehalose, combined with PVA on the survival of S. thermophilus were also explored. Notably,. S. thermophilus stored in 100 mg/mL trehalose and 1 mg/mL PVA +50 mg/mL trehalose combo showed significantly enhanced survival when compared with those in traditional cryoprotectant (20% [v/v] glycerol), which achieved the survival percentage of only 41.03 ± 0.09%. The effects of the freezing temperature and crystallinity on the survival of S. thermophilus were elucidated.
Collapse
Affiliation(s)
- Pei Jiang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Li
- Department of Obstetrics and Gynecology, Shanghai Changhai Hospital, Shanghai, China
| | - Baolin Liu
- Institute of Biothermal and Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Wei Liang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Rolle K, Okotrub KA, Zaytseva IV, Babin SA, Surovtsev NV. Self-pressurised rapid freezing at arbitrary cryoprotectant concentrations. J Microsc 2023; 292:27-36. [PMID: 37615208 DOI: 10.1111/jmi.13220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/17/2023] [Accepted: 08/21/2023] [Indexed: 08/25/2023]
Abstract
Self-pressurised rapid freezing (SPRF) has been proposed as a simple alternative to traditional high-pressure freezing (HPF) protocols for vitrification of biological samples in electron microscopy and cryopreservation applications. Both methods exploit the circumstance that the melting point of ice reaches a minimum when subjected to pressure of around 210 MPa, however, in SPRF its precise quantity depends on sample properties and hence, is generally unknown. In particular, cryoprotective agents (CPAs) are expected to be a factor; though eschewed by many SPRF experiments, vitrification of larger samples notably cannot be envisaged without them. Thus, in this study, we address the question of how CPA concentration affects pressure inside sealed capillaries, and how to design SPRF experiments accordingly. By embedding a fibre-optic probe in samples and performing Raman spectroscopy after freezing, we first present a direct assessment of pressure build-up during SPRF, enabled by the large pressure sensitivity of the Raman shift of hexagonal ice. Choosing dimethyl sulphoxide (DMSO) as a model CPA, this approach allows us to demonstrate that average pressure drops to zero when DMSO concentrations of 15 wt% are exceeded. Since a trade-off between pressure and DMSO concentration represents an impasse with regard to vitrification of larger samples, we introduce a sample architecture with two chambers, separated by a partition that allows for equilibration of pressure but not DMSO concentrations. We show that pressure and concentration in the fibre-facing chamber can be tuned independently, and present differential scanning calorimetry (DSC) data supporting the improved vitrification performance of two-chamber designs. Lay version of abstract for 'Self-pressurised rapid freezing at arbitrary cryoprotectant concentrations' Anyone is familiar with pipes bursting in winter because the volume of ice is greater than that of liquid water. Less well known is the fact that inside a thick-walled container, sealed and devoid of air bubbles, this pressure build-up will allow a fraction of water to remain unfrozen if the sample is also cooled sufficiently rapidly far below the freezing point. This phenomenon has already been harnessed for specimen preparation in microscopy, where low temperatures are useful to immobilise the sample, but harmful if ice formation occurs. However, specimen preparation cannot always rely on this pressure-based effect alone, but sometimes requires addition of chemicals to inhibit ice formation. Not enough is known directly about how these chemicals affect pressure build-up: Indeed, rapid cooling below the freezing point is only possible for small sample volumes, typically placed inside sealed capillaries, so that space is generally insufficient to accommodate a pressure sensor. By means of a compact sensor, based on an optical fibre, laser and spectrometer, we present the first direct assessment of pressure inside sealed capillaries. We show that addition of chemicals reduces pressure build-up and present a two-chambered capillary to circumvent the resulting trade-off. Also, we present evidence showing that the two-chambered capillary design can avoid ice formation more readily than a single-chambered one.
Collapse
Affiliation(s)
- Konrad Rolle
- Institute of Automation and Electrometry SB RAS, Novosibirsk, Russia
| | | | - Irina V Zaytseva
- Institute of Automation and Electrometry SB RAS, Novosibirsk, Russia
| | - Sergei A Babin
- Institute of Automation and Electrometry SB RAS, Novosibirsk, Russia
| | | |
Collapse
|
4
|
Jaskiewicz JJ, Dayao DAE, Girouard D, Sevenler D, Widmer G, Toner M, Tzipori S, Sandlin RD. Scalable cryopreservation of infectious Cryptosporidium hominis oocysts by vitrification. PLoS Pathog 2023; 19:e1011425. [PMID: 37289815 PMCID: PMC10284403 DOI: 10.1371/journal.ppat.1011425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/21/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Cryptosporidium hominis is a serious cause of childhood diarrhea in developing countries. The development of therapeutics is impeded by major technical roadblocks including lack of cryopreservation and simple culturing methods. This impacts the availability of optimized/standardized singular sources of infectious parasite oocysts for research and human challenge studies. The human C. hominis TU502 isolate is currently propagated in gnotobiotic piglets in only one laboratory, which limits access to oocysts. Streamlined cryopreservation could enable creation of a biobank to serve as an oocyst source for research and distribution to other investigators requiring C. hominis. Here, we report cryopreservation of C. hominis TU502 oocysts by vitrification using specially designed specimen containers scaled to 100 μL volume. Thawed oocysts exhibit ~70% viability with robust excystation and 100% infection rate in gnotobiotic piglets. The availability of optimized/standardized sources of oocysts may streamline drug and vaccine evaluation by enabling wider access to biological specimens.
Collapse
Affiliation(s)
- Justyna J. Jaskiewicz
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Children`s Boston, Boston, Massachusetts, United States of America
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Denise Ann E. Dayao
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Donald Girouard
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Derin Sevenler
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Children`s Boston, Boston, Massachusetts, United States of America
| | - Giovanni Widmer
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Mehmet Toner
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Children`s Boston, Boston, Massachusetts, United States of America
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Rebecca D. Sandlin
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Children`s Boston, Boston, Massachusetts, United States of America
| |
Collapse
|
5
|
Cui M, Zhan T, Yang J, Dang H, Yang G, Han H, Liu L, Xu Y. Droplet Generation, Vitrification, and Warming for Cell Cryopreservation: A Review. ACS Biomater Sci Eng 2023; 9:1151-1163. [PMID: 36744931 DOI: 10.1021/acsbiomaterials.2c01087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cryopreservation is currently a key step in translational medicine that could provide new ideas for clinical applications in reproductive medicine, regenerative medicine, and cell therapy. With the advantages of a low concentration of cryoprotectant, fast cooling rate, and easy operation, droplet-based printing for vitrification has received wide attention in the field of cryopreservation. This review summarizes the droplet generation, vitrification, and warming method. Droplet generation techniques such as inkjet printing, microvalve printing, and acoustic printing have been applied in the field of cryopreservation. Droplet vitrification includes direct contact with liquid nitrogen vitrification and droplet solid surface vitrification. The limitations of droplet vitrification (liquid nitrogen contamination, droplet evaporation, gas film inhibition of heat transfer, frosting) and solutions are discussed. Furthermore, a comparison of the external physical field warming method with the conventional water bath method revealed that better applications can be achieved in automated rapid warming of microdroplets. The combination of droplet vitrification technology and external physical field warming technology is expected to enable high-throughput and automated cryopreservation, which has a promising future in biomedicine and regenerative medicine.
Collapse
Affiliation(s)
- Mengdong Cui
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Taijie Zhan
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Jiamin Yang
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Hangyu Dang
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Guoliang Yang
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Hengxin Han
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Linfeng Liu
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| | - Yi Xu
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai200093, China
| |
Collapse
|
6
|
Freitas-Ribeiro S, Reis RL, Pirraco RP. Long-term and short-term preservation strategies for tissue engineering and regenerative medicine products: state of the art and emerging trends. PNAS NEXUS 2022; 1:pgac212. [PMID: 36714838 PMCID: PMC9802477 DOI: 10.1093/pnasnexus/pgac212] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 02/01/2023]
Abstract
There is an ever-growing need of human tissues and organs for transplantation. However, the availability of such tissues and organs is insufficient by a large margin, which is a huge medical and societal problem. Tissue engineering and regenerative medicine (TERM) represent potential solutions to this issue and have therefore been attracting increased interest from researchers and clinicians alike. But the successful large-scale clinical deployment of TERM products critically depends on the development of efficient preservation methodologies. The existing preservation approaches such as slow freezing, vitrification, dry state preservation, and hypothermic and normothermic storage all have issues that somehow limit the biomedical applications of TERM products. In this review, the principles and application of these approaches will be summarized, highlighting their advantages and limitations in the context of TERM products preservation.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal,ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Barco GMR, Portugal
| | - Rui L Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal,ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Barco GMR, Portugal
| | | |
Collapse
|
7
|
Linkova DD, Rubtsova YP, Egorikhina MN. Cryostorage of Mesenchymal Stem Cells and Biomedical Cell-Based Products. Cells 2022; 11:cells11172691. [PMID: 36078098 PMCID: PMC9454587 DOI: 10.3390/cells11172691] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) manifest vast opportunities for clinical use due both to their ability for self-renewal and for effecting paracrine therapeutic benefits. At the same time, difficulties with non-recurrent generation of large numbers of cells due to the necessity for long-term MSC expansion ex vivo, or the requirement for repeated sampling of biological material from a patient significantly limits the current use of MSCs in clinical practice. One solution to these problems entails the creation of a biobank using cell cryopreservation technology. This review is aimed at analyzing and classifying literature data related to the development of protocols for the cryopreservation of various types of MSCs and tissue-engineered structures. The materials in the review show that the existing techniques and protocols for MSC cryopreservation are very diverse, which significantly complicates standardization of the entire process. Here, the selection of cryoprotectors and of cryoprotective media shows the greatest variability. Currently, it is the cryopreservation of cell suspensions that has been studied most extensively, whereas there are very few studies in the literature on the freezing of intact tissues or of tissue-engineered structures. However, even now it is possible to develop general recommendations to optimize the cryopreservation process, making it less traumatic for cells.
Collapse
|
8
|
Ren S, Shu Z, Pan J, Wang Z, Ma R, Peng J, Chen M, Gao D. Single-Mode Electromagnetic Resonance Rewarming for the Cryopreservation of Samples with Large Volumes: A Numerical and Experimental Study. Biopreserv Biobank 2022; 20:317-322. [PMID: 35984939 DOI: 10.1089/bio.2022.0107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rapid and uniform rewarming has been proved to be beneficial, and sometimes indispensable for the survival of cryopreserved biomaterials, inhibiting ice-recrystallization-devitrification and thermal stress-induced fracture (especially in large samples). To date, the convective water bath remains the gold standard rewarming method for small samples in the clinical settings, but it failed in the large samples (e.g., cryopreserved tissues and organs) due to damage caused by the slow and nonuniform heating. A single-mode electromagnetic resonance (SMER) system was developed to achieve ultrafast and uniform rewarming for large samples. In this study, we investigated the heating effects of the SMER system and compared the heating performance with water bath and air warming. A numerical model was established to further analyze the temperature change and distribution at different time points during the rewarming process. Overall, the SMER system achieved rapid heating at 331.63 ± 8.59°C min-1 while limiting the maximum thermal gradient to <9°C min-1, significantly better than the other two warming methods. The experimental results were highly consistent, indicating SMER is a promising rewarming technology for the successful cryopreservation of large biosamples.
Collapse
Affiliation(s)
- Shen Ren
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA.,Department of Mechanical Engineering, Seattle University, Seattle, Washington, USA
| | - Zhiquan Shu
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA.,School of Engineering and Technology, University of Washington-Tacoma, Tacoma, Washington, USA
| | - Jiaji Pan
- College of Engineering and Design, Hunan Normal University, Changsha, Hunan, China.,State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, China
| | - Ziyuan Wang
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| | - Ruidong Ma
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| | - Ji Peng
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| | - Ming Chen
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
9
|
Heydarzadeh S, Kia SK, Boroomand S, Hedayati M. Recent Developments in Cell Shipping Methods. Biotechnol Bioeng 2022; 119:2985-3006. [PMID: 35898166 DOI: 10.1002/bit.28197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/09/2022] [Accepted: 07/17/2022] [Indexed: 11/11/2022]
Abstract
As opposed to remarkable advances in the cell therapy industry, researches reveal inexplicable difficulties associated with preserving and post-thawing cell death. Post cryopreservation apoptosis is a common occurrence that has attracted the attention of scientists to use apoptosis inhibitors. Transporting cells without compromising their survival and function is crucial for any experimental cell-based therapy. Preservation of cells allows the safe transportation of cells between distances and improves quality control testing in clinical and research applications. The vitality of transported cells is used to evaluate the efficacy of transportation strategies. For many decades, the conventional global methods of cell transfer were not only expensive but also challenging and had adverse effects. The first determination of some projects is optimizing cell survival after cryopreservation. The new generation of cryopreservation science wishes to find appropriate and alternative methods for cell transportation to ship viable cells at an ambient temperature without dry ice or in media-filled flasks. The diversity of cell therapies demands new cell shipping methodologies and cryoprotectants. In this review, we tried to summarize novel improved cryopreservation methods and alternatives to cryopreservation with safe and viable cell shipping at ambient temperature, including dry preservation, hypothermic preservation, gel-based methods, encapsulation methods, fibrin microbeads, and osmolyte solution compositions. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shabnam Heydarzadeh
- Department of Biochemistry, School of Biological Sciences, Falavarjan Branch Islamic Azad University, Isfahan, Iran.,Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sima Kheradmand Kia
- Laboratory for Red Blood Cell Diagnostics, Sanquin, Amsterdam, The Netherlands
| | - Seti Boroomand
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mehdi Hedayati
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Kwizera EA, Stewart S, Mahmud MM, He X. Magnetic Nanoparticle-Mediated Heating for Biomedical Applications. JOURNAL OF HEAT TRANSFER 2022; 144:030801. [PMID: 35125512 PMCID: PMC8813031 DOI: 10.1115/1.4053007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/03/2021] [Indexed: 05/17/2023]
Abstract
Magnetic nanoparticles, especially superparamagnetic nanoparticles (SPIONs), have attracted tremendous attention for various biomedical applications. Facile synthesis and functionalization together with easy control of the size and shape of SPIONS to customize their unique properties, have made it possible to develop different types of SPIONs tailored for diverse functions/applications. More recently, considerable attention has been paid to the thermal effect of SPIONs for the treatment of diseases like cancer and for nanowarming of cryopreserved/banked cells, tissues, and organs. In this mini-review, recent advances on the magnetic heating effect of SPIONs for magnetothermal therapy and enhancement of cryopreservation of cells, tissues, and organs, are discussed, together with the non-magnetic heating effect (i.e., high Intensity focused ultrasound or HIFU-activated heating) of SPIONs for cancer therapy. Furthermore, challenges facing the use of magnetic nanoparticles in these biomedical applications are presented.
Collapse
Affiliation(s)
- Elyahb Allie Kwizera
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742
| | - Md Musavvir Mahmud
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
11
|
A simple and efficient cryopreservation method for mouse small intestinal and colon organoids for regenerative medicine. Biochem Biophys Res Commun 2022; 595:14-21. [DOI: 10.1016/j.bbrc.2021.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 11/18/2022]
|
12
|
Uhrig M, Ezquer F, Ezquer M. Improving Cell Recovery: Freezing and Thawing Optimization of Induced Pluripotent Stem Cells. Cells 2022; 11:799. [PMID: 35269421 PMCID: PMC8909336 DOI: 10.3390/cells11050799] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Achieving good cell recovery after cryopreservation is an essential process when working with induced pluripotent stem cells (iPSC). Optimized freezing and thawing methods are required for good cell attachment and survival. In this review, we concentrate on these two aspects, freezing and thawing, but also discuss further factors influencing cell recovery such as cell storage and transport. Whenever a problem occurs during the thawing process of iPSC, it is initially not clear what it is caused by, because there are many factors involved that can contribute to insufficient cell recovery. Thawing problems can usually be solved more quickly when a certain order of steps to be taken is followed. Under optimized conditions, iPSC should be ready for further experiments approximately 4-7 days after thawing and seeding. However, if the freezing and thawing protocols are not optimized, this time can increase up to 2-3 weeks, complicating any further experiments. Here, we suggest optimization steps and troubleshooting options for the freezing, thawing, and seeding of iPSC on feeder-free, Matrigel™-coated, cell culture plates whenever iPSC cannot be recovered in sufficient quality. This review applies to two-dimensional (2D) monolayer cell culture and to iPSC, passaged, frozen, and thawed as cell aggregates (clumps). Furthermore, we discuss usually less well-described factors such as the cell growth phase before freezing and the prevention of osmotic shock during thawing.
Collapse
Affiliation(s)
- Markus Uhrig
- Center for Regenerative Medicine, School of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile;
| | | | - Marcelo Ezquer
- Center for Regenerative Medicine, School of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile;
| |
Collapse
|
13
|
Parwin S, Parui J. Ag nanofluids synthesis in presence of citrate at different stirring rotation and their post reaction stability. J DISPER SCI TECHNOL 2021. [DOI: 10.1080/01932691.2020.1789469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Shama Parwin
- Chemistry Division, School of Advanced Sciences, Vellore Institute of Technology, Chennai, India
| | - Jayanta Parui
- Chemistry Division, School of Advanced Sciences, Vellore Institute of Technology, Chennai, India
| |
Collapse
|
14
|
Liu Y, Lin A, Tiersch TR, Monroe WT. A 3D Printed Vitrification Device for Storage in Cryopreservation Vials. APPLIED SCIENCES (BASEL, SWITZERLAND) 2021; 11:7977. [PMID: 36777919 PMCID: PMC9910574 DOI: 10.3390/app11177977] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sperm cryopreservation by vitrification is a promising approach for small-bodied animals such as zebrafish (Danio rerio). However, most vitrification tools adopted in aquatic research were initially designed for applications other than sperm (such as human embryo freezing) and, thus, pose challenges for adoption to sperm vitrification. Three-dimensional (3D) printing combined with open hardware sharing is an emerging strategy to address challenges in the development of cryopreservation tools. The goal of this study was to develop a 3D printed Vitrification Device for Cryo-Vials (VDCV) that can be integrated with the existing vial storage systems. The VDCV combined the vitrification and handling components to achieve functions of sample handling, vitrification, storage, and identification. The vitrification component featured a base, a stem, and a loop. A total of 36 configurations with various loop lengths (8, 10, and 12 mm); loop widths (2.0, 2.5, 3.0, and 3.5 mm); and support structures (open, transverse, and axial) of the VDCD prototypes were designed, fabricated, and tested. Device handling orientations (horizontal and vertical holding angles prior to and during freezing) were also investigated. Computer simulations estimated that the cooling rate of the samples ranged from 0.6-1.5 × 105 °C/min in all the configurations. Prior to freezing, loops with axial supports produced a minimum of 92% film retention. The overall trends of full vitrification occurrence were observed: horizontal plunging > vertical plunging, and axial support > transverse support and open loop. A loop length of 8 mm had the highest overall vitrification occurrence (86-100%). No significant differences (p = 0.6584) were shown in a volume capacity (5.7-6.0 μL) among the three supporting configurations. A single unit of VDCV can provide loading efficiencies of about 6 × 107 sperm/vial, pooling of samples from 3-6 males/vial, and fertilization for 1800 eggs/vial. The VDCV are low-cost (<$0.5 material cost per unit) and can be customized, standardized, securely labeled, and efficiently stored. The prototypes can be accessed by user communities through open-fabrication file sharing and fabricated with consumer-level 3D printers, thus facilitating community-level standardization.
Collapse
Affiliation(s)
- Yue Liu
- Department of Biological and Agricultural Engineering, Louisiana State University Agricultural Center, Louisiana State University, Baton Rouge, LA 70803, USA
- Aquatic Germplasm and Genetic Resources Center, School of Renewable Natural Resources, Louisiana State University Agricultural Center, Baton Rouge, LA 70820, USA
| | - Andy Lin
- Department of Biological and Agricultural Engineering, Louisiana State University Agricultural Center, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Terrence R. Tiersch
- Aquatic Germplasm and Genetic Resources Center, School of Renewable Natural Resources, Louisiana State University Agricultural Center, Baton Rouge, LA 70820, USA
| | - William Todd Monroe
- Department of Biological and Agricultural Engineering, Louisiana State University Agricultural Center, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
15
|
Huang H, He X, Yarmush ML. Advanced technologies for the preservation of mammalian biospecimens. Nat Biomed Eng 2021; 5:793-804. [PMID: 34426675 PMCID: PMC8765766 DOI: 10.1038/s41551-021-00784-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 06/23/2021] [Indexed: 02/07/2023]
Abstract
The three classical core technologies for the preservation of live mammalian biospecimens-slow freezing, vitrification and hypothermic storage-limit the biomedical applications of biospecimens. In this Review, we summarize the principles and procedures of these three technologies, highlight how their limitations are being addressed via the combination of microfabrication and nanofabrication, materials science and thermal-fluid engineering and discuss the remaining challenges.
Collapse
Affiliation(s)
- Haishui Huang
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, USA.
- Bioinspired Engineering and Biomechanics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States.
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, USA.
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
16
|
In Situ Vitrification of Lung Cancer Organoids on a Microwell Array. MICROMACHINES 2021; 12:mi12060624. [PMID: 34071266 PMCID: PMC8227627 DOI: 10.3390/mi12060624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/15/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Three-dimensional cultured patient-derived cancer organoids (PDOs) represent a powerful tool for anti-cancer drug development due to their similarity to the in vivo tumor tissues. However, the culture and manipulation of PDOs is more difficult than 2D cultured cell lines due to the presence of the culture matrix and the 3D feature of the organoids. In our other study, we established a method for lung cancer organoid (LCO)-based drug sensitivity tests on the superhydrophobic microwell array chip (SMAR-chip). Here, we describe a novel in situ cryopreservation technology on the SMAR-chip to preserve the viability of the organoids for future drug sensitivity tests. We compared two cryopreservation approaches (slow freezing and vitrification) and demonstrated that vitrification performed better at preserving the viability of LCOs. Next, we developed a simple procedure for in situ cryopreservation and thawing of the LCOs on the SMAR-chip. We proved that the on-chip cryopreserved organoids can be recovered successfully and, more importantly, showing similar responses to anti-cancer drugs as the unfrozen controls. This in situ vitrification technology eliminated the harvesting and centrifugation steps in conventional cryopreservation, making the whole freeze–thaw process easier to perform and the preserved LCOs ready to be used for the subsequent drug sensitivity test.
Collapse
|
17
|
Abbasi Y, Hajiaghalou S, Baniasadi F, Mahabadi VP, Ghalamboran MR, Fathi R. Fe 3O 4 magnetic nanoparticles improve the vitrification of mouse immature oocytes and modulate the pluripotent genes expression in derived pronuclear-stage embryos. Cryobiology 2021; 100:81-89. [PMID: 33781804 DOI: 10.1016/j.cryobiol.2021.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 03/20/2021] [Accepted: 03/20/2021] [Indexed: 01/15/2023]
Abstract
The vitrification of Germinal Vesicle (immature) oocytes is beneficial for preservation of fertility in cases involving reproductive problems. The use of nanoparticles (NP(s)) as vitrification aid is a novel approach towards improving vitrification efficiency. The efficacy of use of iron oxide (Fe3O4) nanoparticles as vitrification aid is reported in this paper. Immature oocytes from NMRI mice were collected and divided into non-vitrified (nVit), Vitrified (Vit) and Vitrified + NP (Vit+NP) groups. In the Vit+NP group, solutions containing Fe3O4 nanoparticles at three different concentrations (0.004%, 0.008% and 0.016% w/v) were separately added to the vitrification solution and their effects on the vitrification of the oocytes were compared. The concentration that was found to be best performing (0.004% w/v) was used in vitrification studies in subsequent experiments. Mitochondrial function, apoptosis incidence, ultrastructure alteration, nuclear maturity, embryo formation and genes expression (Nanog, Oct4, Cdx2, and Sox2) were evaluated in response to the addition of the nanoparticle solution during vitrification. Nuclear maturity of oocyte and embryo formation increased significantly (P ≤ 0.05) in the vitrified + NP group. Expression of Sox2 also increased significantly in both vitrified and vitrified + NP groups. While there was a significant increase in Oct4 expression in the vitrified group as compared to control, there was no significant difference between vitrified and Vit+NP groups. The expression of Cdx2 decreased significantly (P ≤ 0.05) in the Vit+NP group. From these observations, Fe3O4 nanoparticles could protect immature oocytes from cryodamages, positively affect vitrification and modulate the pluripotency of derived pronuclear-stage embryos.
Collapse
Affiliation(s)
- Yasaman Abbasi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Department of Cell and Molecular Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Farzaneh Baniasadi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Faculty of Science, Physics Department, Shahid Beheshti University, Iran
| | - Vahid Pirhajati Mahabadi
- Neuroscience Research Center, Iran University of Medical Science, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran
| | | | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
18
|
Abstract
Vitrification is an alternative to cryopreservation by freezing that enables hydrated living cells to be cooled to cryogenic temperatures in the absence of ice. Vitrification simplifies and frequently improves cryopreservation because it eliminates mechanical injury from ice, eliminates the need to find optimal cooling and warming rates, eliminates the importance of differing optimal cooling and warming rates for cells in mixed cell type populations, eliminates the need to find a frequently imperfect compromise between solution effects injury and intracellular ice formation, and can enable chilling injury to be "outrun" by using rapid cooling without a risk of intracellular ice formation. On the other hand, vitrification requires much higher concentrations of cryoprotectants than cryopreservation by freezing, which introduces greater risks of both osmotic damage and cryoprotectant toxicity. Fortunately, a large number of remedies for the latter problem have been discovered over the past 35 years, and osmotic damage can in most cases be eliminated or adequately controlled by paying careful attention to cryoprotectant introduction and washout techniques. Vitrification therefore has the potential to enable the superior and convenient cryopreservation of a wide range of biological systems (including molecules, cells, tissues, organs, and even some whole organisms), and it is also increasingly recognized as a successful strategy for surviving harsh environmental conditions in nature. But the potential of vitrification is sometimes limited by an insufficient understanding of the complex physical and biological principles involved, and therefore a better understanding may not only help to improve present outcomes but may also point the way to new strategies that may be yet more successful in the future. This chapter accordingly describes the basic principles of vitrification and indicates the broad potential biological relevance of this alternative method of cryopreservation.
Collapse
|
19
|
Watanabe H, Akiyama Y. Improved and reproducible cell viability in the superflash freezing method using an automatic thawing apparatus. Cryobiology 2020; 96:12-18. [PMID: 32946776 DOI: 10.1016/j.cryobiol.2020.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/22/2020] [Accepted: 09/11/2020] [Indexed: 11/29/2022]
Abstract
Cell cryopreservation stops the biological activity of cells by placing them in the frozen state, and can be used to preserve cells without subculturing, which can cause contamination and genetic drift. However, the freezing process used in cryopreservation can injure or damage the cells due to the cytotoxicity of cryoprotecting agents (CPAs). We have previously reported a CPA-free cryopreservation method based on inkjet technology. In this method, the vitrified cells were exposed to the room temperature atmosphere during the transport of the cells using tweezers, which caused devitrification due to the increased temperature and often lowered the cell viability. In the present study, we developed an automatic thawing apparatus that transports the vitrified cells rapidly into a prewarmed medium using a spring hinge. Observations with a high-speed camera revealed that the spring hinge drops the cells into the prewarmed medium within 20 ms. All heat-transfer simulations for the apparatuses with different designs and rotation speeds showed that the cells remained below the glass-transition temperature during the transport. Finally, the apparatus was evaluated using mouse fibroblast 3T3 cells. The cell viability was improved and its reproducibility was enhanced using this apparatus. The results indicate that the combination of superflash freezing with the rapid thawing process represents a promising approach to circumvent the problems typically associated with the addition of CPAs.
Collapse
Affiliation(s)
- Hiroki Watanabe
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan
| | - Yoshitake Akiyama
- Graduate School of Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan; Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano, 386-8567, Japan.
| |
Collapse
|
20
|
Sheikhpour M, Arabi M, Kasaeian A, Rokn Rabei A, Taherian Z. Role of Nanofluids in Drug Delivery and Biomedical Technology: Methods and Applications. Nanotechnol Sci Appl 2020; 13:47-59. [PMID: 32801669 PMCID: PMC7399455 DOI: 10.2147/nsa.s260374] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/23/2020] [Indexed: 01/04/2023] Open
Abstract
Recently, suspensions of several nanoparticles or nanocomposites have attained a vast field of application in biomedical research works in some specified conditions and clinical trials. These valuable suspensions, which allow the nanoparticles to disperse and act in homogenous and stable media, are named as nanofluids. Several studies have introduced the advantages of nanofluids in biomedical approaches in different fields. Few review articles have been reported for presenting an overview of the wide biomedical applications of nanofluids, such as diagnosis and therapy. The review is focused on nanosuspensions, as the nanofluids with solid particles. Major applications are focused on nanosuspension, which is the main type of nanofluids. So, concise content about major biomedical applications of nanofluids in drug delivery systems, imaging, and antibacterial activities is presented in this paper. For example, applying magnetic nanofluid systems is an important route for targeted drug delivery, hyperthermia, and differential diagnosis. Also, nanofluids could be used as a potential antibacterial agent to overcome antibiotic resistance. This study could be useful for presenting the novel and applicable methods for success in current medical practice.
Collapse
Affiliation(s)
- Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mohadeseh Arabi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alibakhsh Kasaeian
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Ali Rokn Rabei
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Zahra Taherian
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| |
Collapse
|
21
|
Jaskiewicz JJ, Sevenler D, Swei AA, Widmer G, Toner M, Tzipori S, Sandlin RD. Cryopreservation of infectious Cryptosporidium parvum oocysts achieved through vitrification using high aspect ratio specimen containers. Sci Rep 2020; 10:11711. [PMID: 32678171 PMCID: PMC7366687 DOI: 10.1038/s41598-020-68643-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/16/2020] [Indexed: 01/11/2023] Open
Abstract
Infection with protozoa of the genus Cryptosporidium is a leading cause of child morbidity and mortality associated with diarrhea in the developing world. Research on this parasite has been impeded by many technical limitations, including the lack of cryopreservation methods. While cryopreservation of Cryptosporidium oocysts by vitrification was recently achieved, the method is restricted to small sample volumes, thereby limiting widespread implementation of this procedure. Here, a second-generation method is described for cryopreservation of C. parvum oocysts by vitrification using custom high aspect ratio specimen containers, which enable a 100-fold increase in sample volume compared to previous methods. Oocysts cryopreserved using the described protocol exhibit high viability, maintain in vitro infectivity, and are infectious to interferon-gamma (IFN-γ) knockout mice. Importantly, the course of the infection is comparable to that observed in mice infected with unfrozen oocysts. Vitrification of C. parvum oocysts in larger volumes will expedite progress of research by enabling the sharing of isolates among different laboratories and the standardization of clinical trials.
Collapse
Affiliation(s)
- Justyna J Jaskiewicz
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Derin Sevenler
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children, Boston, MA, USA
| | - Anisa A Swei
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children, Boston, MA, USA
| | - Giovanni Widmer
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children, Boston, MA, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA.
| | - Rebecca D Sandlin
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children, Boston, MA, USA.
| |
Collapse
|
22
|
Xiang X, Liu Z, Zhao G. Sodium Alginate as a Novel Cryoprotective Agent for Cryopreservation of Endothelial Cells in a Closed Polytetrafluoroethylene Loop. Biopreserv Biobank 2020; 18:321-328. [PMID: 32552032 DOI: 10.1089/bio.2020.0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human umbilical vein endothelial cells (HUVECs) have wide applications in tissue engineering, drug delivery, and other fields due to their low antigenicity. Therefore, it is of great significance to effectively cryopreserve HUVECs for subsequent use (i.e., transport, long-term storage of cell banks). However, many commonly used cryoprotective agents (CPAs) are cytotoxic, so finding ideal CPAs to reduce the damage will pave the way for the application of HUVEC's cryopreservation. In this study, sodium alginate (SA) was employed as one of the main CPAs in a closed polytetrafluoroethylene (PTFE) loop used for cryopreservation with fast freezing of HUVECs. The ice crystal growth process was observed and the thermal enthalpy changes and osmolality of different solutions were tested. Moreover, the effects on cell viability and recovery were examined. The results showed that the addition of SA delayed the growth of ice crystals and decreased the number of ice crystals. Specifically, when 0.5% (w/v) SA was added to the CPAs, the cell survival increased by 10%. It is proved in this study that SA can be used as a novel CPA in combination with PTFE for the fast freezing of HUVECs, which is expected to improve the survival rate of cells and promote the exploration of protectants and cryopreservation in the future.
Collapse
Affiliation(s)
- Xingxue Xiang
- Department of Thermal Science and Energy Engineering and University of Science and Technology of China, Hefei, People's Republic of China
| | - Zhifeng Liu
- Department of Thermal Science and Energy Engineering and University of Science and Technology of China, Hefei, People's Republic of China
| | - Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, People's Republic of China
| |
Collapse
|
23
|
Xie X, Diao Z, Cahill DG. Microscale, bendable thermoreflectance sensor for local measurements of the thermal effusivity of biological fluids and tissues. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2020; 91:044903. [PMID: 32357710 DOI: 10.1063/1.5141376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Measurements of the thermal transport properties of biological fluids and tissues are important for biomedical applications such as thermal diagnostics and thermal therapeutics. Here, we describe a microscale thermoreflectance sensor to measure the thermal effusivity of fluids and biological samples in a minimally invasive manner. The sensor is based on ultrafast optical pump-probe techniques and employs a metal-coated optical fiber as both a photonic waveguide and a local probe. Calibration of the sensor with five liquids shows that the percentage deviation between experimentally measured effusivity and literature values is on average <3%. We further demonstrate the capability of the sensor by measuring the thermal effusivity of vegetable oil, butter, pork liver, and quail egg white and yolk. We relate the thermal effusivity of the samples to their composition and water content, and establish our technique as a powerful and flexible method for studying the local thermal transport properties of biological materials.
Collapse
Affiliation(s)
- Xu Xie
- Materials Research Laboratory, Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Zhu Diao
- Materials Research Laboratory, Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - David G Cahill
- Materials Research Laboratory, Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
24
|
Huang H, Rey-Bedón C, Yarmush ML, Usta OB. Deep-supercooling for extended preservation of adipose-derived stem cells. Cryobiology 2020; 92:67-75. [PMID: 31751557 PMCID: PMC7195234 DOI: 10.1016/j.cryobiol.2019.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/04/2023]
Abstract
Cell preservation is an enabling technology for widespread distribution and applications of mammalian cells. Traditional cryopreservation via slow-freezing or vitrification provides long-term storage but requires cytotoxic cryoprotectants (CPA) and tedious CPA loading/unloading, cooling, and recovering procedures. Hypothermic storage around 0-4 °C is an alternative method but only works for a short period due to its high storage temperatures. Here, we report on the deep-supercooling (DSC) preservation of human adipose-derived stem cells at deep subzero temperatures without freezing for extended storage. Enabled by surface sealing with an immiscible oil phase, cell suspension can be preserved in a liquid state at -13 °C and -16 °C for 7 days with high cell viability, retention of stemness, attachment, and multilineage differentiation capacities. These results demonstrate that DSC is an improved short-term preservation approach to provide off-the-shelf cell sources for booming cell-based medicine and bioengineering.
Collapse
Affiliation(s)
- Haishui Huang
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States
| | - Camilo Rey-Bedón
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States; Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, United States.
| | - O Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States.
| |
Collapse
|
25
|
Hou Y, Lu C, Dou M, Zhang C, Chang H, Liu J, Rao W. Soft liquid metal nanoparticles achieve reduced crystal nucleation and ultrarapid rewarming for human bone marrow stromal cell and blood vessel cryopreservation. Acta Biomater 2020; 102:403-415. [PMID: 31734413 DOI: 10.1016/j.actbio.2019.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/01/2019] [Accepted: 11/11/2019] [Indexed: 12/19/2022]
Abstract
High warming rates during cryopreservation are crucial and essential for successful vitrification. However, realizing a faster warming rate in low-concentration cryoprotective agents appears to be challenging for conventional warming process through convective heat transfer. Herein, we developed a liquid metal (LM) nanosystem that can act as a spatial source to significantly enhance the warming rates with near-infrared laser irradiation during the warming process. The synthetic Pluronic F127-liquid metal nanoparticles (PLM NPs) displayed multiple performances with uniform particle size, superior photothermal conversion efficiency (52%), repeatable photothermal stability, and low cytotoxicity. Particularly, it is more difficult for the liquid PLM NPs with less surface free energy to form crystal nucleation than other solid NPs such as gold and Fe3O4, which is beneficial for the cooling process during cryopreservation. The viability of human bone marrow-derived mesenchymal stem cells postcryopreservation reached 78±3%, which is threefold higher than that obtained by the conventional warming method (25±6%). Additionally, the cells postcryopreservation maintained their normal attachment, proliferation, surface marker expression, and intact multilineage differentiation properties. Moreover, the results of mouse tails including blood vessel cryopreservation showed a relatively improved intact structure when using PLM NP rewarming compared with the results of conventional warming. The new LM nanosystem provides a universal platform for cryopreservation that is expected to have potential for widespread applications including bioengineering, cell-based medicine, and clinical translation. STATEMENT OF SIGNIFICANCE: In this study, we fabricated soft liquid metal nanoparticles with high photothermal conversion efficiency, repeatable photothermal stability, and low cytotoxicity. Particularly, soft liquid metal nanoparticles with less surface free energy and suppression effects of ice formation were first introduced to mediate cryopreservation. Superior ice-crystallization inhibition is achieved as a result of less crystal nucleation and ultrarapid rewarming during the freezing and warming processes of cryopreservation, respectively. Collectively, cryopreservation of human bone marrow stromal cells (HBMSCs) and mouse tails including blood vessels can be successfully performed using this new nanoplatform, showing great potential in the application of soft nanoparticles in cryopreservation.
Collapse
|
26
|
Stastna JJ, Yiapanas AD, Mandawala AA, Fowler KE, Harvey SC. Cryopreservation produces limited long-term effects on the nematode Caenorhabditis elegans. Cryobiology 2019; 92:86-91. [PMID: 31770528 DOI: 10.1016/j.cryobiol.2019.11.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 11/19/2022]
Abstract
Cryopreservation, the freezing and later warming of biological samples with minimal loss of viability, is important in many scientific disciplines. For some applications, particularly those where there is limited available material, it is critical to ensure the maximal survival rates of cryopreserved materials. Most of the challenges encountered with such techniques take place after the warming process where cryodamage affects cell viability and future development. Here we have used the nematode Caenorhabditis elegans to investigate the effects of cryodamage caused by slow-freezing. We find that freezing results in the death of some worms, with an approximately 40% reduction in the number of worms that develop in the frozen populations, but that the effects on worms that survive are limited. For example, there are no differences in the lifetime fecundity or in lifespan between frozen and control worms, although early fecundity and body size was reduced in frozen worms. Similarly, analyses of body wall muscle structure and of pharyngeal function indicates that muscle development and function are not significantly affected by freezing. We do however determine that freezing increases the rates of matricidal hatching, where progeny hatch within the mother. Overall, these results indicate that, for worms that survive, cryopreservation produces limited long-term effects, but do indicate that some phenotypes could be used in further analyses of the cellular damage induced by cryopreservation.
Collapse
Affiliation(s)
- J J Stastna
- Biomolecular Research Group, Canterbury Christ Church University, Canterbury, CT1 1QU, UK
| | - A D Yiapanas
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - A A Mandawala
- Biomolecular Research Group, Canterbury Christ Church University, Canterbury, CT1 1QU, UK
| | - K E Fowler
- Biomolecular Research Group, Canterbury Christ Church University, Canterbury, CT1 1QU, UK
| | - S C Harvey
- Biomolecular Research Group, Canterbury Christ Church University, Canterbury, CT1 1QU, UK.
| |
Collapse
|
27
|
A non-traditional approach to cryopreservation by ultra-rapid cooling for human mesenchymal stem cells. PLoS One 2019; 14:e0220055. [PMID: 31329628 PMCID: PMC6645672 DOI: 10.1371/journal.pone.0220055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 07/08/2019] [Indexed: 11/26/2022] Open
Abstract
Cryopreservation is the most common method for long-term cell storage. Successful cryopreservation of cells depends on optimal freezing conditions, freezer storage and a proper thawing technique to minimize the cellular damage that can occur during the cryopreservation process. These factors are especially critical for sensitive stem cells with a consequential and significant impact on viability and functionality. Until now, slow-freezing has been the routine method of cryopreservation but, more recently rapid-cooling techniques have also been proposed. In this study, an ultra-rapid cooling technique [1] was performed for the first time on human mesenchymal stem cells and the effectiveness evaluated in comparison with the conventional slow-freezing procedure. A thin nylon-membrane carrier was used combined with different cryoprotective agents: dimethyl sulfoxide, ethylene glycol and/or trehalose. Various aspects of the low cryoprotective doses and the ultra-rapid cooling procedure of the human mesenchymal stem cells were examined including: the physical properties of the nylon-support, cells encumbrance, viability, proliferation and differentiation. The expression of cell surface markers and apoptosis were also investigated. The study used an ultra-rapid cooling/warming method and showed an overall cell integrity preservation (83–99%), with no significant differences between dimethyl sulfoxide or ethylene glycol treatment (83–87%) and a substantial cell viability of 68% and 51%, respectively. We confirmed a discrepancy also observed by other authors in cell viability and integrity, which implies that caution is necessary when assessing and reporting cell viability data.
Collapse
|
28
|
Khosla K, Zhan L, Bhati A, Carley-Clopton A, Hagedorn M, Bischof J. Characterization of Laser Gold Nanowarming: A Platform for Millimeter-Scale Cryopreservation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7364-7375. [PMID: 30299961 PMCID: PMC6536355 DOI: 10.1021/acs.langmuir.8b03011] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Preventing ice formation during cryopreservation by vitrification has led to the successful storage and banking of numerous cellular- and tissue-based biomaterials. In their breakthrough work, Peter Mazur's group achieved over 90% survival by using a laser warming technique for 100 μm mice oocytes that were cooled in 0.1 μL droplets with 2.3 M CPA and extracellularly loaded India ink (laser absorber). Laser warming can provide rapid and uniform warming rates to "outrun" damaging ice crystal growth. Here we generalize Mazur's technique for microliter-sized droplets using laser nanowarming to rewarm millimeter-scale biomaterials when loaded extracellularly and/or intracellularly with biocompatible 1064 nm resonant gold nanoparticles. First, we show that droplets containing low-concentration cryoprotectants (such as 2 M propylene glycol ± 1 M trehalose) can be rapidly cooled at rates up to 90 000 °C/min by plunging into liquid nitrogen to achieve either a visually transparent state (i.e., vitrified) or a cloudy with ice (i.e., nonvitrified) state. Both modeling and experiments were then used to characterize the laser nanowarming process for different laser energy (2-6 J), pulse length (1-20 ms), droplet volume (0.2-1.8 μL), cryoprotectant (2-3 M), and gold concentration (0.77 × 1017-4.8 × 1017 nps/m3) values to assess physical and biological success. Physical success was achieved by finding conditions that minimize cloudiness and white spots within the droplets during cooling and warming as signs of damaging ice formation and ice crystallization, respectively. Biological success was achieved using human dermal fibroblasts to find conditions that achieve ≥90% cell viability normalized to controls postwarming. Thus, physical and biological success can be achieved using this platform cryopreservation approach of rapid cooling and laser gold nanowarming in millimeter-scale systems.
Collapse
Affiliation(s)
- Kanav Khosla
- Department of Mechanical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55408
| | - Li Zhan
- Department of Mechanical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55408
| | - Aditya Bhati
- Department of Mechanical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55408
| | - Aiden Carley-Clopton
- Department of Mechanical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55408
| | - Mary Hagedorn
- Department of Reproductive Sciences, Smithsonian Conservation Biology Institute, Smithsonian National Zoological Park, Washington DC 20008
- Center for Species Survival, Smithsonian Conservation Biology Institute, Smithsonian National Zoological Park, Washington DC 20008
- Hawaii Institute of Marine Biology, University of Hawaii, 46-007 Lilipuna Road, Kaneohe, Hawaii 96744
| | - John Bischof
- Department of Mechanical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55408
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, MN 55408
| |
Collapse
|
29
|
de Vries RJ, Banik PD, Nagpal S, Weng L, Ozer S, van Gulik TM, Toner M, Tessier SN, Uygun K. Bulk Droplet Vitrification: An Approach to Improve Large-Scale Hepatocyte Cryopreservation Outcome. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7354-7363. [PMID: 30514081 PMCID: PMC6548701 DOI: 10.1021/acs.langmuir.8b02831] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Loss of hepatocyte viability and metabolic function after cryopreservation is still a major issue. Although vitrification is a promising alternative, it has generally been proven to be unsuitable for vitrification of large cell volumes which is required for clinical applications. Here, we propose a novel bulk droplet (3-5 mm diameter) vitrification method which allows high throughput volumes (4 mL/min), while using a low preincubated CPA concentration (15% v/v) to minimize toxicity and loss of cell viability and function. We used rapid (1.25 s) osmotic dehydration to concentrate a low preincubated intracellular CPA concentration ahead of vitrification, without the need of fully equilibrating toxic CPA concentrations. We compared direct postpreservation viability, long-term viability, and metabolic function of bulk droplet vitrified, cryopreserved, and fresh hepatocytes. Simulations and cooling rate measurements confirmed an adequate concentration of the intracellular CPA concentration (up to 8.53 M) after dehydration in combination with high cooling rates (960-1320 °C/min) for successful vitrification. In comparison to cryopreserved hepatocytes, bulk droplet vitrified hepatocytes had a significantly higher viability, directly after preservation and after 1 day in culture. Moreover, bulk droplet vitrified hepatocytes had evidently better morphology and showed significantly higher metabolic activity than cryopreserved hepatocytes in long-term collagen sandwich cultures. In conclusion, we developed a novel bulk droplet vitrification method of which we validated the theoretical background and demonstrated the feasibility to use this method to vitrify large cell volumes. Moreover, we showed that this method results in improved hepatocyte viability and metabolic function as compared to cryopreservation.
Collapse
Affiliation(s)
- Reinier J. de Vries
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
- Department of Surgery, University of Amsterdam, Amsterdam, the Netherlands
| | - Peony D. Banik
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| | - Sonal Nagpal
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| | - Lindong Weng
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| | - Sinan Ozer
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| | | | - Mehmet Toner
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| | - Shannon N. Tessier
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| | - Korkut Uygun
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine, Harvard Medical School, Boston MA, USA
| |
Collapse
|
30
|
Farooq U, Haider Z, Liang XM, Memon K, Hossain SMC, Zheng Y, Xu H, Qadir A, Panhwar F, Dong S, Zhao G, Luo J. Surface-Acoustic-Wave-Based Lab-on-Chip for Rapid Transport of Cryoprotectants across Cell Membrane for Cryopreservation with Significantly Improved Cell Viability. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1805361. [PMID: 30821937 DOI: 10.1002/smll.201805361] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/04/2019] [Indexed: 06/09/2023]
Abstract
Cryopreservation is essential to effectively extend the shelf life of delicate biomaterials while maintaining proper levels of cell functions. Cryopreservation requires a cryoprotective agent (CPA) to suppress intracellular ice formation during freezing, but it must be removed prior to clinical use due to its toxicity. Conventional multistep CPA loading and unloading approaches are time consuming, often creating osmotic shocks and causing mechanical injuries for biological samples. An efficient surface-acoustic-wave- (SAW-) based lab-on-a-chip (LoC) for fast loading and removal of CPAs is presented here. With the SAW-based multistep CPA loading/removal approach, high concentration (3 m) CPA can be successfully loaded and removed in less than 1 min. Results show that the technique causes the least harm to umbilical cord matrix mesenchymal stem cells as compared to conventional method, and an average of 24% higher cell recovery rate is achieved, while preserving the integrity and morphology of the cells. This device is the first of its kind to combine high loading/unloading efficiency, high cell viability, and high throughput into one LoC device, offering not only a more efficient and safer route for CPA loading and removal from cells, but also paving the way for other cryopreservation-dependent applications.
Collapse
Affiliation(s)
- Umar Farooq
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Zeeshan Haider
- Centre for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Xin M Liang
- Department of Medicine, Department of Veterans Affairs, Division of Hematology and Oncology, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kashan Memon
- Centre for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - S M Chapal Hossain
- Centre for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Yuan Zheng
- Centre for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Hongsheng Xu
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Akeel Qadir
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Fazil Panhwar
- Centre for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Shurong Dong
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Gang Zhao
- Centre for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Jikui Luo
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
- Institute of Renewable Energy & Environmental Technologies, University of Bolton, Bolton, BL3 5AB, UK
| |
Collapse
|
31
|
Clulow J, Upton R, Trudeau VL, Clulow S. Amphibian Assisted Reproductive Technologies: Moving from Technology to Application. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1200:413-463. [PMID: 31471805 DOI: 10.1007/978-3-030-23633-5_14] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amphibians have experienced a catastrophic decline since the 1980s driven by disease, habitat loss, and impacts of invasive species and face ongoing threats from climate change. About 40% of extant amphibians are under threat of extinction and about 200 species have disappeared completely. Reproductive technologies and biobanking of cryopreserved materials offer technologies that could increase the efficiency and effectiveness of conservation programs involving management of captive breeding and wild populations through reduced costs, better genetic management and reduced risk of species extinctions. However, there are relatively few examples of applications of these technologies in practice in on-the-ground conservation programs, and no example that we know of where genetic diversity has been restored to a threatened amphibian species in captive breeding or in wild populations using cryopreserved genetic material. This gap in the application of technology to conservation programs needs to be addressed if assisted reproductive technologies (ARTs) and biobanking are to realise their potential in amphibian conservation. We review successful technologies including non-invasive gamete collection, IVF and sperm cryopreservation that work well enough to be applied to many current conservation programs. We consider new advances in technology (vitrification and laser warming) of cryopreservation of aquatic embryos of fish and some marine invertebrates that may help us to overcome factors limiting amphibian oocyte and embryo cryopreservation. Finally, we address two case studies that illustrate the urgent need and the opportunity to implement immediately ARTs, cryopreservation and biobanking to amphibian conservation. These are (1) managing the biosecurity (disease risk) of the frogs of New Guinea which are currently free of chytridiomycosis, but are at high risk (2) the Sehuencas water frog of Bolivia, which until recently had only one known surviving male.
Collapse
Affiliation(s)
- J Clulow
- School of Environmental and Life Sciences, University of Newcastle, Newcastle, NSW, Australia.
| | - R Upton
- School of Environmental and Life Sciences, University of Newcastle, Newcastle, NSW, Australia
| | - V L Trudeau
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - S Clulow
- Department of Biological Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
32
|
Cao Y, Zhao G, Panhwar F, Zhang X, Chen Z, Cheng L, Zang C, Liu F, Zhao Y, He X. The Unusual Properties of Polytetrafluoroethylene Enable Massive-Volume Vitrification of Stem Cells with Low-Concentration Cryoprotectants. ADVANCED MATERIALS TECHNOLOGIES 2019; 4:1800289. [PMID: 31448319 PMCID: PMC6707752 DOI: 10.1002/admt.201800289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Indexed: 05/13/2023]
Abstract
Injectable stem cell-hydrogel constructs hold great potential for regenerative medicine and cell-based therapies. However, their clinical application is still challenging due to their short shelf-life at ambient temperature and the time-consuming fabrication procedure. Banking the constructs at cryogenic temperature may offer the possibility of "off-the-shelf" availability to end-users. However, ice formation during the cryopreservation process may compromise the construct quality and cell viability. Vitrification, cooling biological samples without apparent ice formation, has been explored to resolve the challenge. However, contemporary vitrification methods are limited to very small volume (up to ~0.25 ml) and/or need highly toxic and high concentration (up to ~8 M) of permeable cryoprotectants (pCPAs). Here, we show that polytetrafluoroethylene (PTFE, best known as Teflon for making non-stick cookware) capillary is flexible and unusually stable at a cryogenic temperature. By using the PTFE capillary as a flexible cryopreservation vessel together with alginate hydrogel microencapsulation and Fe3O4 nanoparticle-mediated nanowarming to suppress ice formation, massive-volume (10 ml) vitrification of cell-alginate hydrogel constructs with a low concentration (~2.5 M) of pCPA can be achieved. This may greatly facilitate the use of stem cell-based constructs for tissue regeneration and cell based therapies in the clinic.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Fazil Panhwar
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Xiaozhang Zhang
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Zhongrong Chen
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Lin Cheng
- Department of Emergency Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Chuanbao Zang
- Yinfeng Cryomedicine Technology Co., LTD, Jinan, China
| | - Feng Liu
- Yinfeng Cryomedicine Technology Co., LTD, Jinan, China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
33
|
Huebinger J. Modification of cellular membranes conveys cryoprotection to cells during rapid, non-equilibrium cryopreservation. PLoS One 2018; 13:e0205520. [PMID: 30304023 PMCID: PMC6179263 DOI: 10.1371/journal.pone.0205520] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/26/2018] [Indexed: 12/05/2022] Open
Abstract
Rapid cooling and re-warming has been shown promising to cryopreserve living cells, which cannot be preserved by conventional slow freezing methods. However, success is limited by the cytotoxicity of highly concentrated cryoprotective agents. Recent results have shown that cryoprotective agents do not need to suppress intracellular ice crystals completely to allow for survival after cryopreservation. Cryoprotective agents like DMSO or ethylene glycol can also lead to a tolerance of cells towards intracellular ice. It is however unclear by which mechanism this tolerance is achieved. These substances are also known to modulate properties of cellular membranes. It is shown here that cryoprotective DMSO and ethylene glycol have a clear influence on the mobility of lipids in the plasma membrane of HeLa cells. To isolate changes of the properties of plasma membranes from effects on ice formation, the membrane properties were modulated in absence of cryoprotective agents. This was achieved by changing their sterol content. In cells with elevated sterol content, an immobile lipid fraction was present, similar to cells treated with DMSO and ethylene glycol. These cells showed also significantly increased plasma membrane integrity after rapid freezing and thawing in the absence of classical cryoprotective agents. However, their intracellular lysosomes, which cannot be enriched with sterols, still got ruptured. These results clearly indicate that a modulation of membrane properties can convey cryoprotection. Upon slow cooling, elevated sterol content had actually an adverse effect on the plasma membranes, which shows that this effect is specific for rapid, non-equilibrium cooling processes. Unraveling this alternative mode of action of cryoprotection should help in the directed design of novel cryoprotective agents, which might be less cytotoxic than classical, empirically-found cryoprotective agents.
Collapse
Affiliation(s)
- Jan Huebinger
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- * E-mail:
| |
Collapse
|
34
|
Huebinger J, Grabenbauer M. Self-Pressurized Rapid Freezing as Cryo-Fixation Method for Electron Microscopy and Cryopreservation of Living Cells. ACTA ACUST UNITED AC 2018; 79:e47. [PMID: 29924483 DOI: 10.1002/cpcb.47] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Reduction or complete prevention of ice crystal formation during freezing of biological specimens is mandatory for two important biological applications: (1) cryopreservation of living cells or tissues for long-term storage, and (2) cryo-fixation for ultrastructural investigations by electron microscopy. Here, a protocol that is fast, easy-to-use, and suitable for both cryo-fixation and cryopreservation is described. Samples are rapidly cooled in tightly sealed metal tubes of high thermal diffusivity and then plunged into a liquid cryogen. Due to the fast cooling speed and high-pressure buildup internally in the confined volume of the metal tubes, ice crystal formation is reduced or completely prevented, resulting in vitrification of the sample. For cryopreservation, however, a similar principle applies to prevent ice crystal formation during re-warming. A detailed description of procedures for cooling (and re-warming) of biological samples using this technique is provided. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Jan Huebinger
- Department of Systemic Cell Biology, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany
| | - Markus Grabenbauer
- Institute for Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
35
|
Liu X, Zhao G, Chen Z, Panhwar F, He X. Dual Suppression Effect of Magnetic Induction Heating and Microencapsulation on Ice Crystallization Enables Low-Cryoprotectant Vitrification of Stem Cell-Alginate Hydrogel Constructs. ACS APPLIED MATERIALS & INTERFACES 2018; 10:16822-16835. [PMID: 29688697 PMCID: PMC6054798 DOI: 10.1021/acsami.8b04496] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Stem cells microencapsulated in hydrogel as stem cell-hydrogel constructs have wide applications in the burgeoning cell-based medicine. Due to their short shelf life at ambient temperature, long-term storage or banking of the constructs is essential to the "off-the-shelf" ready availability needed for their widespread applications. As a high-efficiency, easy-to-operate, low-toxicity, and low-cost method for long-term storage of the constructs, low-cryoprotectant (CPA) vitrification has attracted tremendous attention recently. However, we found many cells in the stem cell-alginate constructs (∼500 μm in diameter) could not attach to the substrate post low-CPA vitrification with ∼2 M penetrating CPAs. To address this problem, we introduced nanowarming via magnetic induction heating (MIH) of Fe3O4 nanoparticles to minimize recrystallization and devitrification during the warming step of the low-CPA vitrification procedure. Our results indicate that high-quality stem cell-alginate hydrogel constructs with an intact microstructure, high immediate cell survival (>80%), and greatly improved attachment efficiency (by nearly three times, 68% versus 24%) of the encapsulated cells could be obtained post-cryopreservation with nanowarming. Moreover, the cells encapsulated in the cell-hydrogel constructs post-cryopreservation maintained normal proliferation under 3D culture and retained intact biological function of multilineage differentiation. This novel low-CPA vitrification approach for cell cryopreservation enabled by the combined use of alginate hydrogel microencapsulation and Fe3O4 nanoparticles-mediated nanowarming may be valuable in facilitating the widespread application of stem cells in the clinic.
Collapse
Affiliation(s)
- Xiaoli Liu
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Zhongrong Chen
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Fazil Panhwar
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
36
|
Manuchehrabadi N, Gao Z, Zhang J, Ring HL, Shao Q, Liu F, McDermott M, Fok A, Rabin Y, Brockbank KGM, Garwood M, Haynes CL, Bischof JC. Improved tissue cryopreservation using inductive heating of magnetic nanoparticles. Sci Transl Med 2017; 9:9/379/eaah4586. [PMID: 28251904 DOI: 10.1126/scitranslmed.aah4586] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/17/2016] [Accepted: 02/09/2017] [Indexed: 12/15/2022]
Abstract
Vitrification, a kinetic process of liquid solidification into glass, poses many potential benefits for tissue cryopreservation including indefinite storage, banking, and facilitation of tissue matching for transplantation. To date, however, successful rewarming of tissues vitrified in VS55, a cryoprotectant solution, can only be achieved by convective warming of small volumes on the order of 1 ml. Successful rewarming requires both uniform and fast rates to reduce thermal mechanical stress and cracks, and to prevent rewarming phase crystallization. We present a scalable nanowarming technology for 1- to 80-ml samples using radiofrequency-excited mesoporous silica-coated iron oxide nanoparticles in VS55. Advanced imaging including sweep imaging with Fourier transform and microcomputed tomography was used to verify loading and unloading of VS55 and nanoparticles and successful vitrification of porcine arteries. Nanowarming was then used to demonstrate uniform and rapid rewarming at >130°C/min in both physical (1 to 80 ml) and biological systems including human dermal fibroblast cells, porcine arteries and porcine aortic heart valve leaflet tissues (1 to 50 ml). Nanowarming yielded viability that matched control and/or exceeded gold standard convective warming in 1- to 50-ml systems, and improved viability compared to slow-warmed (crystallized) samples. Last, biomechanical testing displayed no significant biomechanical property changes in blood vessel length or elastic modulus after nanowarming compared to untreated fresh control porcine arteries. In aggregate, these results demonstrate new physical and biological evidence that nanowarming can improve the outcome of vitrified cryogenic storage of tissues in larger sample volumes.
Collapse
Affiliation(s)
- Navid Manuchehrabadi
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhe Gao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jinjin Zhang
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hattie L Ring
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.,Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN 55455, USA
| | - Qi Shao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Feng Liu
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael McDermott
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alex Fok
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yoed Rabin
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Kelvin G M Brockbank
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA.,Tissue Testing Technologies LLC, North Charleston, SC 29406, USA
| | - Michael Garwood
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Radiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christy L Haynes
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA. .,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
37
|
Zhang Y, Zhao G, Chapal Hossain SM, He X. Modeling and experimental studies of enhanced cooling by medical gauze for cell cryopreservation by vitrification. INTERNATIONAL JOURNAL OF HEAT AND MASS TRANSFER 2017; 114:1-7. [PMID: 29398719 PMCID: PMC5794028 DOI: 10.1016/j.ijheatmasstransfer.2017.06.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Vitrification is considered as an important alternative approach to traditional slow freezing method for cryopreservation of cells. A typical cell vitrification procedure involves a non-equilibrium cooling process commonly accomplished in liquid nitrogen, while in which film boiling is believed to greatly hinder heat transfer surrounding the sample, resulting in incomplete vitrification or a much higher critical concentration. In this study, we developed a simple while effective approach, wrapping traditional French-type straw with medical gauze, to greatly enhance convective heat transfer during cooling by suppress film boiling. We further established a coupled heat transfer model for cooling and warming of cell suspensions to investigate the inherent thermodynamic mechanism in this approach. The model describes both the macroscale thermal distributions in extracellular solution and the microscale ice crystallization inside the cells. The simulation indicated that straws wrapped with medical gauze would increase cell survival subject to vitrification cryopreservation by significantly increasing the cooling rate to inhibit intracellular ice formation (IIF). Our experiments on human umbilical vein endothelial cells (HUVECs) further confirmed the predictions in that the cell survival rate was significantly increased by wrapping straws with medical gauze.
Collapse
Affiliation(s)
- Yuntian Zhang
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - S. M. Chapal Hossain
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, USA
| |
Collapse
|
38
|
Khosla K, Wang Y, Hagedorn M, Qin Z, Bischof J. Gold Nanorod Induced Warming of Embryos from the Cryogenic State Enhances Viability. ACS NANO 2017; 11:7869-7878. [PMID: 28702993 DOI: 10.1021/acsnano.7b02216] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Zebrafish embryos can attain a stable cryogenic state by microinjection of cryoprotectants followed by rapid cooling, but the massive size of the embryo has consistently led to failure during the convective warming process. Here we address this zebrafish cryopreservation problem by using gold nanorods (GNRs) to assist in the warming process. Specifically, we microinjected the cryoprotectant propylene glycol into zebrafish embryos along with GNRs, and the samples were cooled at a rate of 90 000 °C/min in liquid nitrogen. We demonstrated the ability to unfreeze the zebrafish rapidly (1.4 × 107 °C/min) by irradiating the sample with a 1064 nm laser pulse for 1 ms due to the excitation of GNRs. This rapid warming process led to the outrunning of ice formation, which can damage the embryos. The results from 14 trials (n = 223) demonstrated viable embryos with consistent structure at 1 h (31%) and continuing development at 3 h (17%) and movement at 24 h (10%) postwarming. This compares starkly with 0% viability, structure, or movement at all time points in convectively warmed controls (n = 50, p < 0.001, ANOVA). Our nanoparticle-based warming process could be applied to the storage of fish, and with proper modification, can potentially be used for other vertebrate embryos.
Collapse
Affiliation(s)
- Kanav Khosla
- Department of Mechanical Engineering, University of Minnesota at Twin Cities , 111 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Yiru Wang
- Department of Mechanical Engineering, University of Minnesota at Twin Cities , 111 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Mary Hagedorn
- Smithsonian Conservation Biology Institute, Smithsonian National Zoological Park , 3001 Connecticut Avenue NW, Washington, D.C. 20008, United States
- Hawaii Institute of Marine Biology, University of Hawaii , 46-007 Lilipuna Road, Kaneohe, Hawaii 96744, United States
| | - Zhenpeng Qin
- Department of Mechanical Engineering, University of Minnesota at Twin Cities , 111 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - John Bischof
- Department of Mechanical Engineering, University of Minnesota at Twin Cities , 111 Church Street SE, Minneapolis, Minnesota 55455, United States
- Department of Biomedical Engineering, University of Minnesota at Twin Cities , 312 Church Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
39
|
Elliott GD, Wang S, Fuller BJ. Cryoprotectants: A review of the actions and applications of cryoprotective solutes that modulate cell recovery from ultra-low temperatures. Cryobiology 2017; 76:74-91. [DOI: 10.1016/j.cryobiol.2017.04.004] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 04/07/2017] [Accepted: 04/16/2017] [Indexed: 02/08/2023]
|
40
|
Puschmann E, Selden C, Butler S, Fuller B. Liquidus Tracking: Large scale preservation of encapsulated 3-D cell cultures using a vitrification machine. Cryobiology 2017; 76:65-73. [DOI: 10.1016/j.cryobiol.2017.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 04/15/2017] [Accepted: 04/19/2017] [Indexed: 12/28/2022]
|
41
|
Zhao G, Fu J. Microfluidics for cryopreservation. Biotechnol Adv 2017; 35:323-336. [PMID: 28153517 PMCID: PMC6236673 DOI: 10.1016/j.biotechadv.2017.01.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 11/19/2022]
Abstract
Cryopreservation has utility in clinical and scientific research but implementation is highly complex and includes labor-intensive cell-specific protocols for the addition/removal of cryoprotective agents and freeze-thaw cycles. Microfluidic platforms can revolutionize cryopreservation by providing new tools to manipulate and screen cells at micro/nano scales, which are presently difficult or impossible with conventional bulk approaches. This review describes applications of microfluidic tools in cell manipulation, cryoprotective agent exposure, programmed freezing/thawing, vitrification, and in situ assessment in cryopreservation, and discusses achievements and challenges, providing perspectives for future development.
Collapse
Affiliation(s)
- Gang Zhao
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230027, Anhui, PR China.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
42
|
Reversible Cryopreservation of Living Cells Using an Electron Microscopy Cryo-Fixation Method. PLoS One 2016; 11:e0164270. [PMID: 27711254 PMCID: PMC5053471 DOI: 10.1371/journal.pone.0164270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/22/2016] [Indexed: 02/01/2023] Open
Abstract
Rapid cooling of aqueous solutions is a useful approach for two important biological applications: (I) cryopreservation of cells and tissues for long-term storage, and (II) cryofixation for ultrastructural investigations by electron and cryo-electron microscopy. Usually, both approaches are very different in methodology. Here we show that a novel, fast and easy to use cryofixation technique called self-pressurized rapid freezing (SPRF) is–after some adaptations–also a useful and versatile technique for cryopreservation. Sealed metal tubes with high thermal diffusivity containing the samples are plunged into liquid cryogen. Internal pressure builds up reducing ice crystal formation and therefore supporting reversible cryopreservation through vitrification of cells. After rapid rewarming of pressurized samples, viability rates of > 90% can be reached, comparable to best-performing of the established rapid cooling devices tested. In addition, the small SPRF tubes allow for space-saving sample storage and the sealed containers prevent contamination from or into the cryogen during freezing, storage, or thawing.
Collapse
|
43
|
Zheng Y, Zhao G, Panhwar F, He X. Vitreous Cryopreservation of Human Umbilical Vein Endothelial Cells with Low Concentration of Cryoprotective Agents for Vascular Tissue Engineering. Tissue Eng Part C Methods 2016; 22:964-973. [PMID: 27673413 PMCID: PMC5079420 DOI: 10.1089/ten.tec.2016.0335] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/26/2016] [Indexed: 01/16/2023] Open
Abstract
Cryopreservation of human umbilical vein endothelial cells (HUVECs) is important to tissue engineering applications and the study of the role of endothelial cells in cardiovascular and cerebrovascular diseases. The traditional methods for cryopreservation by vitrification (cooling samples to a cryogenic temperature without apparent freezing) using high concentration of cryoprotective agents (CPAs) and slow freezing are suboptimal due to the severe toxicity of high concentration of CPAs and ice formation-induced cryoinjuries, respectively. In this study, we developed a method to cryopreserve HUVECs by vitrification with low concentration of CPAs. This is achieved by optimizing the CPAs and using highly thermally conductive quartz capillary (QC) to contain samples for vitrification. The latter minimizes the thermal mass to create ultra-fast cooling/warming rates. Our data demonstrate that HUVECs can be vitrified in the QC using 1.4 mol/L ethylene glycol and 1.1 mol/L dimethyl sulfoxide with more than 90% viability. Moreover, this method significantly improves the attachment efficiency of the cryopreserved HUVECs. The attached cells post-cryopreservation proliferate similarly to fresh cells. Therefore, this study may provide an effective vitrification technique to bank HUVECs for vascular tissue engineering and other applications.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Gang Zhao
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
- Anhui Provincial Engineering Research Center for Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Fazil Panhwar
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| |
Collapse
|
44
|
Scheiner D, Bracone G, Imesch P, Fink D, Hehl J, Imthurn B. Comparison of quartz vials with polypropylene vials for rapid cryopreservation of human ovarian tissue. J Ovarian Res 2016; 9:59. [PMID: 27670300 PMCID: PMC5037623 DOI: 10.1186/s13048-016-0268-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 09/17/2016] [Indexed: 11/23/2022] Open
Abstract
Background Because higher survival of follicles during the freezing/thawing procedure improves the quality of cryopreserved tissue reimplanted after oncological therapies, defining an optimal method for human ovarian tissue cryopreservation remains a major issue in this field. One option to improve the cryopreservation procedure is to use better materials, i.e., vials with better conductivity. The aim of this study was to compare polypropylene (PP) with quartz vials. Between September 2012 and January 2013, eight patients were recruited. The ovarian cortex was cut into 3 slices, assigned randomly to a fresh and a cryopreserved group in PP (method B) or quartz vials (method C). Histological and immunohistochemical (IHC) analysis were used. For IHC three antibodies were analyzed: Ki67 (proliferation index), Bcl2 (anti apoptotic index) and Hsp70 (stress index). Results The majority of GCs showed positive staining for Bcl2 in both cryopreservation device, with higher expression in group C than in group B. Oocytes and their nuclei showed intense positive staining for ki67 in both methods B and C, and also a patch positive stromal cells staining for Ki67. Expression of hsp70 was not increased after cryopreservation. Conclusions Cryopreservation using quartz vials led to larger numbers of good follicles while maintaining consistent preservation for stromal cells and vessels.
Collapse
Affiliation(s)
- D Scheiner
- Department of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland.
| | - G Bracone
- Kantonsspital Luzern, Neue Frauenklinik - Kinderwunsch Zentrum, Andrologie-IVF Labor, Spitalstrasse 2, 6000, Luzern, Switzerland
| | - P Imesch
- Department of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland
| | - D Fink
- Department of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland
| | - J Hehl
- LMSC-Light Microscopy and Sreening Centre, ETH Zurich, Schafmattstrasse 18, 8093, Zurich, Switzerland
| | - B Imthurn
- Division of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland
| |
Collapse
|
45
|
Gallardo M, Hebles M, Migueles B, Dorado M, Aguilera L, González M, Piqueras P, Montero L, Sánchez-Martín P, Sánchez-Martín F, Risco R. Thermal and clinical performance of a closed device designed for human oocyte vitrification based on the optimization of the warming rate. Cryobiology 2016; 73:40-6. [PMID: 27312427 DOI: 10.1016/j.cryobiol.2016.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 11/27/2022]
Abstract
Although it was qualitatively pointed out by Fahy et al. (1984), the key role of the warming rates in non-equillibrium vitrification has only recently been quantitatively established for murine oocytes by Mazur and Seki (2011). In this work we study the performance of a closed vitrification device designed under the new paradigm, for the vitrification of human oocytes. The vitrification carrier consists of a main straw in which a specifically designed capillary is mounted and where the oocytes are loaded by aspiration. It can be hermetically sealed before immersion in liquid nitrogen for vitrification, and it is warmed in a sterile water bath at 37 °C. Measured warming rates achieved with this design were of 600.000 ºC/min for a standard DMEM solution and 200.000 ºC/min with the vitrification solution for human oocytes. A cohort of 143 donor MII sibling human oocytes was split into two groups: control (fresh) and vitrified with SafeSpeed device. Similar results were found in both groups: survival (97.1%), fertilization after ICSI (74.7% in control vs. 77.3% in vitrified) and good quality embryos at day three (54.3% in control vs. 58.1% in vitrified) were settled as performance indicators. The pregnancy rate was 3/6 (50%) for the control, 2/3 (66%) for vitrified and 4/5 (80%) for mixed transfers.
Collapse
Affiliation(s)
- Miguel Gallardo
- University of Seville, C/ Camino de los Descubrimientos s/n, 41092, Seville, Spain; Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - María Hebles
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - Beatriz Migueles
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - Mónica Dorado
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - Laura Aguilera
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - Mercedes González
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - Paloma Piqueras
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | - Lorena Montero
- Ginemed Clínicas, C/ Farmacéutico Murillo Herrera nº 3-5, 41010, Seville, Spain
| | | | | | - Ramón Risco
- University of Seville, C/ Camino de los Descubrimientos s/n, 41092, Seville, Spain; National Accelerator Centre, C/ Thomas Alva Edison 7, 41092, Seville, Spain.
| |
Collapse
|
46
|
Daniele N, Campus M, Pellegrini C, Shkembi E, Zinno F. Biobanks and Clinical Research: An "Interesting" Connection. ACTA ACUST UNITED AC 2016. [DOI: 10.17352/acp.000005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
47
|
Magnetic induction heating of superparamagnetic nanoparticles during rewarming augments the recovery of hUCM-MSCs cryopreserved by vitrification. Acta Biomater 2016; 33:264-74. [PMID: 26802443 DOI: 10.1016/j.actbio.2016.01.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 12/24/2015] [Accepted: 01/19/2016] [Indexed: 01/03/2023]
Abstract
Cryopreservation by vitrification has been recognized as a promising strategy for long-term banking of living cells. However, the difficulty to generate a fast enough heating rate to minimize devitrification and recrystallization-induced intracellular ice formation during rewarming is one of the major obstacles to successful vitrification. We propose to overcome this hurdle by utilizing magnetic induction heating (MIH) of magnetic nanoparticles to enhance rewarming. In this study, superparamagnetic (SPM) Fe3O4 nanoparticles were synthesized by a chemical coprecipitation method. We successfully applied the MIH of Fe3O4 nanoparticles for rewarming human umbilical cord matrix mesenchymal stem cells (hUCM-MSCs) cryopreserved by vitrification. Our results show that extracellular Fe3O4 nanoparticles with MIH may efficiently suppress devitrification and/or recrystallization during rewarming and significantly improve the survival of vitrified cells. We further optimized the concentration of Fe3O4 nanoparticles and the current of an alternating current (AC) magnetic field for generating the MIH to maximize cell viability. Our results indicate that MIH in an AC magnetic field with 0.05% (w/v) Fe3O4 nanoparticles significantly facilitates rewarming and improves the cryopreservation outcome of hUCM-MSCs by vitrification. The application of MIH of SPM nanoparticles to achieve rapid and spatially homogeneous heating is a promising strategy for enhanced cryopreservation of stem cells by vitrification. STATEMENT OF SIGNIFICANCE Here we report the successful synthesis and application of Fe3O4 nanoparticles for magnetic induction heating (MIH) to enhance rewarming of vitrification-cryopreserved human umbilical cord matrix mesenchymal stem cells (hUCM-MSCs). We found that MIH-enhanced rewarming greatly improves the survival of vitrification-cryopreserved hUCM-MSCs. Moreover, the hUCM-MSCs retain their intact stemness and multilineage potential of differentiation post cryopreservation by vitrification with the MIH-enhanced rewarming. Therefore, the novel MIH-enhanced cell vitrification is valuable to facilitate the long-term storage of hUCM-MSCs and possibly many other important cells to meet their ever-increasing demand by the burgeoning cell-based medicine.
Collapse
|
48
|
Shi M, Ling K, Yong KW, Li Y, Feng S, Zhang X, Pingguan-Murphy B, Lu TJ, Xu F. High-Throughput Non-Contact Vitrification of Cell-Laden Droplets Based on Cell Printing. Sci Rep 2015; 5:17928. [PMID: 26655688 PMCID: PMC4677291 DOI: 10.1038/srep17928] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/09/2015] [Indexed: 11/09/2022] Open
Abstract
Cryopreservation is the most promising way for long-term storage of biological samples e.g., single cells and cellular structures. Among various cryopreservation methods, vitrification is advantageous by employing high cooling rate to avoid the formation of harmful ice crystals in cells. Most existing vitrification methods adopt direct contact of cells with liquid nitrogen to obtain high cooling rates, which however causes the potential contamination and difficult cell collection. To address these limitations, we developed a non-contact vitrification device based on an ultra-thin freezing film to achieve high cooling/warming rate and avoid direct contact between cells and liquid nitrogen. A high-throughput cell printer was employed to rapidly generate uniform cell-laden microdroplets into the device, where the microdroplets were hung on one side of the film and then vitrified by pouring the liquid nitrogen onto the other side via boiling heat transfer. Through theoretical and experimental studies on vitrification processes, we demonstrated that our device offers a high cooling/warming rate for vitrification of the NIH 3T3 cells and human adipose-derived stem cells (hASCs) with maintained cell viability and differentiation potential. This non-contact vitrification device provides a novel and effective way to cryopreserve cells at high throughput and avoid the contamination and collection problems.
Collapse
Affiliation(s)
- Meng Shi
- School of Energy and Power Engineering, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| | - Kai Ling
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,State Key Laboratory of Mechanical Structure Strength and Vibration, School of Aerospace, Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| | - Kar Wey Yong
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Yuhui Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| | - Shangsheng Feng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,State Key Laboratory of Mechanical Structure Strength and Vibration, School of Aerospace, Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| | - Xiaohui Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,State Key Laboratory of Mechanical Structure Strength and Vibration, School of Aerospace, Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P.R. China.,MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China
| |
Collapse
|
49
|
Huang H, Choi JK, Rao W, Zhao S, Agarwal P, Zhao G, He X. Alginate Hydrogel Microencapsulation Inhibits Devitrification and Enables Large-Volume Low-CPA Cell Vitrification. ADVANCED FUNCTIONAL MATERIALS 2015; 25:6939-6850. [PMID: 26640426 PMCID: PMC4667367 DOI: 10.1002/adfm.201503047] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cryopreservation of stem cells is important to meet their ever-increasing demand by the burgeoning cell-based medicine. The conventional slow freezing for stem cell cryopreservation suffers from inevitable cell injury associated with ice formation and the vitrification (i.e., no visible ice formation) approach is emerging as a new strategy for cell cryopreservation. A major challenge to cell vitrification is intracellular ice formation (IIF, a lethal event to cells) induced by devitrification (i.e., formation of visible ice in previously vitrified solution) during warming the vitrified cells at cryogenic temperature back to super-zero temperatures. Consequently, high and toxic concentrations of penetrating cryoprotectants (i.e., high CPAs, up to ~8 M) and/or limited sample volumes (up to ~2.5 μl) have been used to minimize IIF during vitrification. We reveal that alginate hydrogel microencapsulation can effectively inhibit devitrification during warming. Our data show that if ice formation were minimized during cooling, IIF is negligible in alginate hydrogel-microencapsulated cells during the entire cooling and warming procedure of vitrification. This enables vitrification of pluripotent and multipotent stem cells with up to ~4 times lower concentration of penetrating CPAs (up to 2 M, low CPA) in up to ~100 times larger sample volume (up to ~250 μl, large volume).
Collapse
Affiliation(s)
- Haishui Huang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Department of Mechanical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jung Kyu Choi
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Wei Rao
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Shuting Zhao
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Pranay Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gang Zhao
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA. Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
50
|
Irdani T, Fortunato A, Torre R. An ultra-rapid cryo-technique for complex organisms. Cryobiology 2015; 71:391-7. [PMID: 26499841 DOI: 10.1016/j.cryobiol.2015.10.144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
Abstract
The soil nematode Caenorhabditis elegans is an excellent research model in cell biology, human disease and developmental studies. In this study, a novel cryopreservation technique based on a rapid cooling procedure, previously established for juveniles, was applied to adult-worms. Here we demonstrated that adults of C. elegans, a complex metazoan organism, survive to a rapid cooling and storage in liquid nitrogen (-196 °C) with a very high survival percentage (85%). The procedure relies on a Low CryoProtectant Technique (LCPT) and Ultra Rapid Cooling (URC). The high cooling rate is achieved through the reduction of sample volumes and the effectiveness of a nylon carrier. Our technique complies with the requirements for vitrification to occur. The main distinctive characters of this cryopreservation technique compared to other methods, e.g. Slow Freezing and Vitrification, are presented. Our results show that this cryopreservation method is valid for both unicellular and multicellular organisms; it is suitable for short or long time storage in liquid-nitrogen. This technique promises to be a unique and simpler method for cryostorage of cells, organisms and tissues.
Collapse
Affiliation(s)
- T Irdani
- CRA Consiglio per la Ricerca in Agricoltura e l'Analisi dell'Economia Agraria, 50125 Firenze, Italy.
| | - A Fortunato
- Department of Surgery, University of California, San Francisco, CA, USA and Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| | - R Torre
- European Laboratory for Non-linear Spectroscopy (LENS) and Dipartimento di Fisica e Astronomia, Università degli Studi di Firenze, 50019 Sesto Fiorentino, Italy.
| |
Collapse
|