1
|
Liu X, Zhang L, Li H, Yang J, Zhang L. The Inhibition of Interfacial Ice Formation and Stress Accumulation with Zwitterionic Betaine and Trehalose for High-Efficiency Skin Cryopreservation. RESEARCH (WASHINGTON, D.C.) 2024; 7:0520. [PMID: 39545039 PMCID: PMC11561590 DOI: 10.34133/research.0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/17/2024]
Abstract
Cryopreservation is a promising technique for the long-term storage of skin. However, the formation of ice crystals during cryopreservation unavoidably damages skin structure and functionality. Currently, the lack of thorough and systematic investigation into the internal mechanisms of skin cryoinjury obstructs the advancement of cryopreservation technology. In this study, we identified 3 primary contributors to skin cryoinjury: interfacial ice nucleation, stress accumulation, and thermal stress escalation. We emphasized the paramount role of interfacial ice nucleation in provoking ice growth within the skin during the cooling process. This progress subsequently leads to stress accumulation within the skin. During the rewarming process, the brittleness of skin, previously subjected to freezing, experienced a marked increase in thermal stress due to ice recrystallization. Based on these insights, we developed a novel zwitterionic betaine-based solution formulation designed for cryopreservation skin. This cryoprotective agent formulation exhibited superior capability in lowering ice nucleation temperatures and inhibiting ice formation at interfaces, while also facilitating the growth of smooth and rounded ice crystals compared to sharp-edged and cornered crystals formed in aqueous solutions. As a result, we successfully achieved prolonged cryopreservation of the skin for at least 6 months, while preserving 98.7% of structural integrity and 94.7% of Young's modulus. This work provides valuable insights into the mechanisms of ice crystal damage during organ cryopreservation and profoundly impacts the field of organ transplantation and regenerative medicine.
Collapse
Affiliation(s)
- Xinmeng Liu
- Department of Biochemical Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300350, China
| | - Liming Zhang
- Department of Biochemical Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300350, China
| | - Haoyue Li
- Department of Biochemical Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300350, China
| | - Jing Yang
- Department of Biochemical Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300350, China
| | - Lei Zhang
- Department of Biochemical Engineering, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300350, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
2
|
Khaydukova IV, Ivannikova VM, Zhidkov DA, Belikov NV, Peshkova MA, Timashev PS, Tsiganov DI, Pushkarev AV. Current State and Challenges of Tissue and Organ Cryopreservation in Biobanking. Int J Mol Sci 2024; 25:11124. [PMID: 39456905 PMCID: PMC11508709 DOI: 10.3390/ijms252011124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Recent years have witnessed significant advancements in the cryopreservation of various tissues and cells, yet several challenges persist. This review evaluates the current state of cryopreservation, focusing on contemporary methods, notable achievements, and ongoing difficulties. Techniques such as slow freezing and vitrification have enabled the successful preservation of diverse biological materials, including embryos and ovarian tissue, marking substantial progress in reproductive medicine and regenerative therapies. These achievements highlight improved post-thaw survival and functionality of cryopreserved samples. However, there are remaining challenges such as ice crystal formation, which can lead to cell damage, and the cryopreservation of larger, more complex tissues and organs. This review also explores the role of cryoprotectants and the importance of optimizing both cooling and warming rates to enhance preservation outcomes. Future research priorities include developing new cryoprotective agents, elucidating the mechanisms of cryoinjury, and refining protocols for preserving complex tissues and organs. This comprehensive overview underscores the transformative potential of cryopreservation in biomedicine, while emphasizing the necessity for ongoing innovation to address existing challenges.
Collapse
Affiliation(s)
- Irina V. Khaydukova
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Valeria M. Ivannikova
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Dmitry A. Zhidkov
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Nikita V. Belikov
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Maria A. Peshkova
- Institute for Regenerative Medicine, Sechenov University, 119048 Moscow, Russia
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 119048 Moscow, Russia
| | - Dmitry I. Tsiganov
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
| | - Aleksandr V. Pushkarev
- Department of Refrigeration and Cryogenic Technology, Conditioning Systems, and Life Support Systems, Bauman Moscow State Technical University, 105005 Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
| |
Collapse
|
3
|
Barra JM, Kratz AT, Castro-Gutierrez R, Proia J, Bhardwaj G, Phelps EA, Russ HA. Cryopreservation of Stem Cell-Derived β-Like Cells Enriches for Insulin-Producing Cells With Improved Function. Diabetes 2024; 73:1687-1696. [PMID: 39083654 PMCID: PMC11417432 DOI: 10.2337/db24-0346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/07/2024] [Indexed: 08/02/2024]
Abstract
The generation of stem cell-derived β-like cells (sBCs) holds promise as not only an abundant insulin-producing cell source for replacement therapy of type 1 diabetes (T1D) but also as an invaluable model system for investigating human β-cell development, immunogenicity, and function. Several groups have developed methodology to direct differentiate human pluripotent stem cells into pancreatic cell populations that include glucose-responsive sBCs. Nevertheless, the process of generating sBCs poses substantial experimental challenges. It involves lengthy differentiation periods, there is substantial variability in efficiency, and there are inconsistencies in obtaining functional sBCs. Here, we describe a simple and effective cryopreservation approach for sBC cultures that yields homogeneous sBC clusters that are enriched for insulin-expressing cells while simultaneously depleting proliferative progenitors. Thawed sBCs have enhanced glucose-stimulated insulin release compared with controls in vitro and can effectively engraft and function in vivo. Collectively, this approach alleviates current challenges with inefficient and variable sBC generation while improving their functional state. We anticipate that these findings can inform ongoing clinical application of sBCs for the treatment of patients with T1D and serve as an important resource for the wider diabetes field that will allow for accelerated research discoveries. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | | | - James Proia
- Diabetes Institute, University of Florida, Gainesville, FL
| | | | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Holger A. Russ
- Diabetes Institute, University of Florida, Gainesville, FL
| |
Collapse
|
4
|
Sahoo A, Das PK, Dasu VV, Patra S. Insulin evolution: A holistic view of recombinant production advancements. Int J Biol Macromol 2024; 277:133951. [PMID: 39032893 DOI: 10.1016/j.ijbiomac.2024.133951] [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: 04/23/2024] [Revised: 06/29/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
The increased prevalence of diabetes and the growing popularity of non-invasive methods of recombinant human insulin uptake, such as oral insulin, have increased insulin demand, further limiting the affordability of insulin. Over 40 years have passed since the development of engineered microorganisms that replaced the animal pancreas as the primary source of insulin. To stay ahead of the need for insulin in the present and the future, a few drawbacks with the existing expression systems need to be alleviated, including the inclusion body formation, the use of toxic inducers, and high process costs. To address these bottlenecks and improve insulin production, a variety of techniques are being used in bacteria, yeasts, transgenic plants and animals, mammalian cell lines, and cell-free expression systems. Different approaches for the production of insulin, including two-chain, proinsulin or mini-proinsulin, preproinsulin coupled with fusion protein, chaperone, signal peptide, and purification tags, are explored in upstream, whereas downstream processing takes into account the recovery of intact protein in its bioactive form and purity. This article focuses on the strategies used in the upstream and downstream phases of the bioprocess to produce recombinant human insulin. This review also covers a range of analytical methods and tools employed in investigating the genuity of recombinant human insulin.
Collapse
Affiliation(s)
- Ansuman Sahoo
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 781039, Assam, India
| | - Prabir Kumar Das
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 781039, Assam, India
| | - Veeranki Venkata Dasu
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 781039, Assam, India.
| | - Sanjukta Patra
- Enzyme & Microbial Technology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 781039, Assam, India
| |
Collapse
|
5
|
Pan J, Zeng Q, Peng K, Zhou Y, Shu Z. Review of Rewarming Methods for Cryopreservation. Biopreserv Biobank 2024; 22:304-311. [PMID: 37751240 DOI: 10.1089/bio.2023.0015] [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: 09/27/2023] Open
Abstract
Cryopreservation is the most effective technology for the long-term preservation of biological materials, including cells, tissues, and even organs in the future. The process of cooling and rewarming is essential to the successful preservation of biological materials. One of the critical problems in the development of cryopreservation is the optimization of effective rewarming technologies. This article reviewed rewarming methods, including traditional boundary rewarming commonly used for small-volume biological materials and other advanced techniques that could be potentially feasible for organ preservation in the future. The review focused on various rewarming technique principles, typical applications, and their possible limitations for cryopreservation of biological materials. This article introduced nanowarming methods in the progressing optimization and the possible difficulties. The trends of novel rewarming methods were discussed, and suggestions were given for future development.
Collapse
Affiliation(s)
- Jiaji Pan
- Department of Mechanical Engineering, College of Engineering and Design, Hunan Normal University, Changsha, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Qijin Zeng
- Department of Mechanical Engineering, College of Engineering and Design, Hunan Normal University, Changsha, China
| | - Ke Peng
- Department of Mechanical Engineering, College of Engineering and Design, Hunan Normal University, Changsha, China
| | - Yulin Zhou
- Shuda College, Hunan Normal University, Changsha, China
| | - Zhiquan Shu
- School of Engineering and Technology, University of Washington, Tacoma, Washington, USA
| |
Collapse
|
6
|
Verhoeff K, Cuesta-Gomez N, Maghera J, Dadheech N, Pawlick R, Smith N, O'Gorman D, Razavy H, Marfil-Garza B, Young LG, Thiesen A, MacDonald PE, Shapiro AMJ. Scalable Bioreactor-based Suspension Approach to Generate Stem Cell-derived Islets From Healthy Donor-derived iPSCs. Transplantation 2024:00007890-990000000-00819. [PMID: 39024165 DOI: 10.1097/tp.0000000000005108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) offer the potential to generate autologous iPSC-derived islets (iPSC islets), however, remain limited by scalability and product safety. METHODS Herein, we report stagewise characterization of cells generated following a bioreactor-based differentiation protocol. Cell characteristics were assessed using flow cytometry, quantitative reverse transcription polymerase chain reaction, patch clamping, functional assessment, and in vivo functional and immunohistochemistry evaluation. Protocol yield and costs are assessed to determine scalability. RESULTS Differentiation was capable of generating 90.4% PDX1+/NKX6.1+ pancreatic progenitors and 100% C-peptide+/NKX6.1+ iPSC islet cells. However, 82.1%, 49.6%, and 0.9% of the cells expressed SOX9 (duct), SLC18A1 (enterochromaffin cells), and CDX2 (gut cells), respectively. Explanted grafts contained mature monohormonal islet-like cells, however, CK19+ ductal tissues persist. Using this protocol, semi-planar differentiation using 150 mm plates achieved 5.72 × 104 cells/cm2 (total 8.3 × 106 cells), whereas complete suspension differentiation within 100 mL Vertical-Wheel bioreactors significantly increased cell yield to 1.1 × 106 cells/mL (total 105.0 × 106 cells), reducing costs by 88.8%. CONCLUSIONS This study offers a scalable suspension-based approach for iPSC islet differentiation within Vertical-Wheel bioreactors with thorough characterization of the ensuing product to enable future protocol comparison and evaluation of approaches for off-target cell elimination. Results suggest that bioreactor-based suspension differentiation protocols may facilitate scalability and clinical implementation of iPSC islet therapies.
Collapse
Affiliation(s)
- Kevin Verhoeff
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nerea Cuesta-Gomez
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Jasmine Maghera
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Nidheesh Dadheech
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Rena Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Nancy Smith
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Doug O'Gorman
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Haide Razavy
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Braulio Marfil-Garza
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
- CHRISTUS-LatAm Hub-Excellence and Innovation Center, Monterrey, Mexico
| | | | - Aducio Thiesen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Patrick E MacDonald
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - A M James Shapiro
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
7
|
Shin DY, Park JS, Lee HS, Shim W, Jin L, Lee KW, Park JB, Kim DH, Kim JH. The effect of hydroxyethyl starch as a cryopreservation agent during freezing of mouse pancreatic islets. Biochem Biophys Rep 2024; 38:101658. [PMID: 38362049 PMCID: PMC10867579 DOI: 10.1016/j.bbrep.2024.101658] [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: 11/08/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024] Open
Abstract
Islet transplantation is the most effective treatment strategy for type 1 diabetes. Long-term storage at ultralow temperatures can be used to prepare sufficient islets of good quality for transplantation. For freezing islets, dimethyl sulfoxide (DMSO) is a commonly used penetrating cryoprotective agent (CPA). However, the toxicity of DMSO is a major obstacle to cell cryopreservation. Hydroxyethyl starch (HES) has been proposed as an alternative CPA. To investigate the effects of two types of nonpermeating CPA, we compared 4 % HES 130 and HES 200 to 10 % DMSO in terms of mouse islet yield, viability, and glucose-stimulated insulin secretion (GSIS). After one day of culture, islets were cryopreserved in each solution. After three days of cryopreservation, islet recovery was significantly higher in the HES 130 and HES 200 groups than in the DMSO group. Islet viability in the HES 200 group was also significantly higher than that in the DMSO group on Day 1 and Day 3. Stimulation indices determined by GSIS were higher in the HES 130 and 200 groups than in the DMSO group on Day 3. After three days of cryopreservation, HES 130 and HES 200 both reduced the expression of apoptosis- and necrosis-associated proteins and promoted the survival of islets. In conclusion, the use of HES as a CPA improved the survival and insulin secretion of cryopreserved islets compared with the use of a conventional CPA.
Collapse
Affiliation(s)
- Du Yeon Shin
- Transplantation Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Graduate School, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Jae Suh Park
- Department of Pediatrics, Hematology/Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
| | - Han-Sin Lee
- R&D Center, Cellstormer, Suwon-si, Gyeonggi-do, 16677, Republic of Korea
| | - Wooyoung Shim
- R&D Center, Cellstormer, Suwon-si, Gyeonggi-do, 16677, Republic of Korea
| | - Lauren Jin
- Department of Pediatrics, Hematology/Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
| | - Kyo Won Lee
- Transplantation Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Jae Berm Park
- Transplantation Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Graduate School, Sungkyunkwan University, Seoul, 06351, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Dong Hyun Kim
- Department of Pediatrics, Hematology/Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
| | - Jae Hyeon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Graduate School, Sungkyunkwan University, Seoul, 06351, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06355, Republic of Korea
| |
Collapse
|
8
|
Wang Y, Huang R, Lu Y, Liu M, Mo R. Immuno-protective vesicle-crosslinked hydrogel for allogenic transplantation. Nat Commun 2024; 15:5176. [PMID: 38890279 PMCID: PMC11189436 DOI: 10.1038/s41467-024-49135-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
The longevity of grafts remains a major challenge in allogeneic transplantation due to immune rejection. Systemic immunosuppression can impair graft function and can also cause severe adverse effects. Here, we report a local immuno-protective strategy to enhance post-transplant persistence of allografts using a mesenchymal stem cell membrane-derived vesicle (MMV)-crosslinked hydrogel (MMV-Gel). MMVs are engineered to upregulate expression of Fas ligand (FasL) and programmed death ligand 1 (PD-L1). The MMVs are retained within the hydrogel by crosslinking. The immuno-protective microenvironment of the hydrogel protects allografts by presenting FasL and PD-L1. The binding of these ligands to T effector cells, the dominant contributors to graft destruction and rejection, results in apoptosis of T effector cells and generation of regulatory T cells. We demonstrate that implantation with MMV-Gel prolongs the survival and function of grafts in mouse models of allogeneic pancreatic islet cells and skin transplantation.
Collapse
Affiliation(s)
- Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Mingqi Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
9
|
Fan Z, Chen Y, Yang Z, Niu Y, Liang K, Zhang Y, Zeng J, Feng Y, Zhang Y, Liu Y, Lv C, Zhao P, Zhou L, Kong W, Li W, Chen H, Han D, Du Y. Superimposed Electric Field Enhanced Electrospray for High-Throughput and Consistent Cell Encapsulation. Adv Healthc Mater 2024:e2400780. [PMID: 38850154 DOI: 10.1002/adhm.202400780] [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: 02/28/2024] [Revised: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Cell encapsulation technology, crucial for advanced biomedical applications, faces challenges in existing microfluidic and electrospray methods. Microfluidic techniques, while precise, can damage vulnerable cells, and conventional electrospray methods often encounter instability and capsule breakage during high-throughput encapsulation. Inspired by the transformation of the working state from unstable dripping to stable jetting triggered by local electric potential, this study introduces a superimposed electric field (SEF)-enhanced electrospray method for cell encapsulation, with improved stability and biocompatibility. Utilizing stiffness theory, the stability of the electrospray, whose stiffness is five times stronger under conical confinement, is quantitatively analyzed. The SEF technique enables rapid, continuous production of ≈300 core-shell capsules per second in an aqueous environment, significantly improving cell encapsulation efficiency. This method demonstrates remarkable potential as exemplified in two key applications: (1) a 92-fold increase in human-derived induced pluripotent stem cells (iPSCs) expansion over 10 d, outperforming traditional 2D cultures in both growth rate and pluripotency maintenance, and (2) the development of liver capsules for steatosis modeling, exhibiting normal function and biomimetic lipid accumulation. The SEF-enhanced electrospray method presents a significant advancement in cell encapsulation technology. It offers a more efficient, stable, and biocompatible approach for clinical transplantation, drug screening, and cell therapy.
Collapse
Affiliation(s)
- Zejun Fan
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Yihan Chen
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yudi Niu
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Kaini Liang
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yan Zhang
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jianan Zeng
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yiting Feng
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuying Zhang
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ye Liu
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, China
| | - Cheng Lv
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peng Zhao
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lv Zhou
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenyu Kong
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjing Li
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haoke Chen
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dongbo Han
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
- National Key Laboratory of Kidney Diseases, Beijing, 100000, China
| |
Collapse
|
10
|
Kavian S, Powell-Palm MJ. Limits of pressure-based ice detection during isochoric vitrification. Cryobiology 2024; 115:104905. [PMID: 38759911 DOI: 10.1016/j.cryobiol.2024.104905] [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: 11/03/2023] [Revised: 04/10/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Vitrification under isochoric (constant-volume or volumetrically confined) conditions has emerged as an intriguing new cryopreservation modality, but the physical complexities of the process confound straight-forward interpretation of experimental results. In particular, the signature pressure-based ice detection used in many isochoric techniques becomes paradoxical during vitrification, wherein the emergence of a sharp increase in pressure reliably indicates the presence of ice, but avoidance of this increase does not necessarily indicate its absence. This phenomenon arises from the rich interplay between thermochemical and thermovolumetric effects in isochoric systems, and muddies efforts to confirm the degree to which a sample has vitrified. In this work, we seek to aid interpretation of isochoric vitrification experiments by calculating thermodynamic limits on the maximum amount of ice that may form without being detected by pressure, and by clarifying the myriad physical processes at play. Neglecting kinetic effects, we develop a simplified thermodynamic model accounting for thermal contraction, cavity formation, ice growth, solute ripening, and glass formation, we evaluate it for a range of chamber materials and solution compositions, and we validate against the acutely limited data available. Our results provide both counter-intuitive insights- lower-concentration solutions may contract less while producing more pressure-undetectable ice growth for example- and a general phenomenological framework by which to evaluate the process of vitrification in isochoric systems. We anticipate that the model herein will enable design of future isochoric protocols with minimized risk of pressure-undetectable ice formation, and provide a thermodynamic foundation from which to build an increasingly rigorous multi-physics understanding of isochoric vitrification.
Collapse
Affiliation(s)
- Soheil Kavian
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, 77803, USA.
| | - Matthew J Powell-Palm
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, 77803, USA; Department of Materials Science and Engineering, Texas A&M University, College Station, TX, 77803, USA; Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77803, USA.
| |
Collapse
|
11
|
Li J, Sun L, Bian F, Pandol SJ, Li L. Emerging approaches for the development of artificial islets. SMART MEDICINE 2024; 3:e20230042. [PMID: 39188698 PMCID: PMC11235711 DOI: 10.1002/smmd.20230042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/05/2024] [Indexed: 08/28/2024]
Abstract
The islet of Langerhans, functioning as a "mini organ", plays a vital role in regulating endocrine activities due to its intricate structure. Dysfunction in these islets is closely associated with the development of diabetes mellitus (DM). To offer valuable insights for DM research and treatment, various approaches have been proposed to create artificial islets or islet organoids with high similarity to natural islets, under the collaborative effort of biologists, clinical physicians, and biomedical engineers. This review investigates the design and fabrication of artificial islets considering both biological and tissue engineering aspects. It begins by examining the natural structures and functions of native islets and proceeds to analyze the protocols for generating islets from stem cells. The review also outlines various techniques used in crafting artificial islets, with a specific focus on hydrogel-based ones. Additionally, it provides a concise overview of the materials and devices employed in the clinical applications of artificial islets. Throughout, the primary goal is to develop artificial islets, thereby bridging the realms of developmental biology, clinical medicine, and tissue engineering.
Collapse
Affiliation(s)
- Jingbo Li
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| | - Lingyu Sun
- Department of Clinical LaboratoryNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Feika Bian
- Department of Clinical LaboratoryNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Stephen J. Pandol
- Division of GastroenterologyDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Ling Li
- Department of EndocrinologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
12
|
Liang H, Zhang R, Zhou L, Wu X, Chen J, Li X, Chen J, Shan L, Wang H. Corn stigma ameliorates hyperglycemia in zebrafish and GK rats of type 2 diabetes. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117746. [PMID: 38216098 DOI: 10.1016/j.jep.2024.117746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cornstigma (CS), derived from the stigma and style of gramineous plant Zeamays. The medicinal use of CS can be traced back to DianNanMateriaMedica. LingnanMedicinalPlantsCompendium records its effectiveness in ameliorating diabetes. Diabetes is a metabolic disorder characterized by hyperglycemia and the consequent chronic complications of kidney, heart, brain and other organs, which pose a significant threat to human health. CS has shown great potential in relieving hyperglycemia associated with diabetes. However, the mechanism of CS in treating diabetes remains unclear. AIM OF THE STUDY To explore the pathogenesis of diabetes and the mechanism of CS improving hyperglycemia in diabetes. MATERIALS AND METHODS We measured apigenin and luteolin contents in CS by UPLC/MS/MS method. Selecting Wistar rats as normal group, and GK rats as model group. For rats, we detected glucose and lipid metabolism indicators, including GHb, AST, ALT, U-Glu, UA, U-TP, U-ALB, and ACR after treatment. For zebrafish, we utilized alloxan and sucrose to establish the diabetes model. Measuring zebrafish blood glucose is employed to evaluate the hypoglycemic capability of CS. In order to explore the mechanism of CS in treating diabetes, we sequenced the transcriptome of zebrafish, compared differentially expressed genes of normal, diabetic, and CS-treated group, and validated multiple enrichment pathways by PCR. RESULTS CS can improve blood glucose levels in both GK rats and diabetic zebrafish. For rats, CS partially restored glucose and lipid metabolism indicators. Transcriptome data from zebrafish showed a close correlation with steroid biosynthesis. The RNA-Sequencing was consistent with PCR results, indicating that CS downregulated gene (fdft1,lss,cyp51) expression concerned with steroid biosynthesis pathway in the diabetes model. CONCLUSION CS effectively improved blood glucose levels, regulated glucose and lipid metabolism by suppressing gene expression in steroid biosynthesis pathway, and ameliorated hyperglycemia. Our research provides valuable insights for CS in the treatment of diabetes, and proposes a new strategy for selecting clinical medications for diabetes.
Collapse
Affiliation(s)
- Haowei Liang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ruiqin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Li Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xiaolong Wu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jingan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xinyue Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jieqiong Chen
- Office of Educational Administration, Zhejiang University of Science and Technology, Hangzhou, China.
| | - Letian Shan
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Hui Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Scientific Research Department, Zhejiang Chinese Medical University, Hangzhou, China; Jinhua Academy, Zhejiang Chinese Medical University, Jinhua, China.
| |
Collapse
|
13
|
Ye Z, Tai Y, Han Z, Liu S, Etheridge ML, Pasek-Allen JL, Shastry C, Liu Y, Li Z, Chen C, Wang Z, Bischof JC, Nam J, Yin Y. Engineering Magnetic Nanoclusters for Highly Efficient Heating in Radio-Frequency Nanowarming. NANO LETTERS 2024; 24:4588-4594. [PMID: 38587406 DOI: 10.1021/acs.nanolett.4c00721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Effective thawing of cryopreserved samples requires rapid and uniform heating. This is achievable through nanowarming, an approach that heats magnetic nanoparticles by using alternating magnetic fields. Here we demonstrate the synthesis and surface modification of magnetic nanoclusters for efficient nanowarming. Magnetite (Fe3O4) nanoclusters with an optimal diameter of 58 nm exhibit a high specific absorption rate of 1499 W/g Fe under an alternating magnetic field at 43 kA/m and 413 kHz, more than twice that of commercial iron oxide cores used in prior nanowarming studies. Surface modification with a permeable resorcinol-formaldehyde resin (RFR) polymer layer significantly enhances their colloidal stability in complex cryoprotective solutions, while maintaining their excellent heating capacity. The Fe3O4@RFR nanoparticles achieved a high average heating rate of 175 °C/min in cryopreserved samples at a concentration of 10 mg Fe/mL and were successfully applied in nanowarming porcine iliac arteries, highlighting their potential for enhancing the efficacy of cryopreservation.
Collapse
Affiliation(s)
- Zuyang Ye
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Youyi Tai
- Department of Bioengineering, University of California, Riverside, California 92521, United States
| | - Zonghu Han
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Sangmo Liu
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Michael L Etheridge
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jacqueline L Pasek-Allen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Chaitanya Shastry
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Yun Liu
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Zhiwei Li
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Chen Chen
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Zhongxiang Wang
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jin Nam
- Department of Bioengineering, University of California, Riverside, California 92521, United States
| | - Yadong Yin
- Department of Chemistry, University of California, Riverside, California 92521, United States
| |
Collapse
|
14
|
Zhang X, Ge L, Jin G, Liu Y, Yu Q, Chen W, Chen L, Dong T, Miyagishima KJ, Shen J, Yang J, Lv G, Xu Y, Yang Q, Ye L, Yi S, Li H, Zhang Q, Chen G, Liu W, Yang Y, Li W, Ou J. Cold-induced FOXO1 nuclear transport aids cold survival and tissue storage. Nat Commun 2024; 15:2859. [PMID: 38570500 PMCID: PMC10991392 DOI: 10.1038/s41467-024-47095-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Cold-induced injuries severely limit opportunities and outcomes of hypothermic therapies and organ preservation, calling for better understanding of cold adaptation. Here, by surveying cold-altered chromatin accessibility and integrated CUT&Tag/RNA-seq analyses in human stem cells, we reveal forkhead box O1 (FOXO1) as a key transcription factor for autonomous cold adaptation. Accordingly, we find a nonconventional, temperature-sensitive FOXO1 transport mechanism involving the nuclear pore complex protein RANBP2, SUMO-modification of transporter proteins Importin-7 and Exportin-1, and a SUMO-interacting motif on FOXO1. Our conclusions are supported by cold survival experiments with human cell models and zebrafish larvae. Promoting FOXO1 nuclear entry by the Exportin-1 inhibitor KPT-330 enhances cold tolerance in pre-diabetic obese mice, and greatly prolongs the shelf-life of human and mouse pancreatic tissues and islets. Transplantation of mouse islets cold-stored for 14 days reestablishes normoglycemia in diabetic mice. Our findings uncover a regulatory network and potential therapeutic targets to boost spontaneous cold adaptation.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Cancer Biology, Dana-Farber Cancer Institute; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Lihao Ge
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Guanghui Jin
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yasong Liu
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
| | - Qingfen Yu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weizhao Chen
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liang Chen
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tao Dong
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kiyoharu J Miyagishima
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Juan Shen
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China
| | - Jinghong Yang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guo Lv
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China
| | - Yan Xu
- Cell-gene Therapy Translational Medicine Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Yang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
| | - Linsen Ye
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuhong Yi
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
| | - Hua Li
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China
- Cell-gene Therapy Translational Medicine Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guihua Chen
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China
| | - Wei Liu
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China.
| | - Yang Yang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China.
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jingxing Ou
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.
- Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong province engineering laboratory for transplantation medicine, Guangzhou, China.
| |
Collapse
|
15
|
Choi J, Cayabyab F, Perez H, Yoshihara E. Scaling Insulin-Producing Cells by Multiple Strategies. Endocrinol Metab (Seoul) 2024; 39:191-205. [PMID: 38572534 PMCID: PMC11066437 DOI: 10.3803/enm.2023.1910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 04/05/2024] Open
Abstract
In the quest to combat insulin-dependent diabetes mellitus (IDDM), allogenic pancreatic islet cell therapy sourced from deceased donors represents a significant therapeutic advance. However, the applicability of this approach is hampered by donor scarcity and the demand for sustained immunosuppression. Human induced pluripotent stem cells are a game-changing resource for generating synthetic functional insulin-producing β cells. In addition, novel methodologies allow the direct expansion of pancreatic progenitors and mature β cells, thereby circumventing prolonged differentiation. Nevertheless, achieving practical reproducibility and scalability presents a substantial challenge for this technology. As these innovative approaches become more prominent, it is crucial to thoroughly evaluate existing expansion techniques with an emphasis on their optimization and scalability. This manuscript delineates these cutting-edge advancements, offers a critical analysis of the prevailing strategies, and underscores pivotal challenges, including cost-efficiency and logistical issues. Our insights provide a roadmap, elucidating both the promises and the imperatives in harnessing the potential of these cellular therapies for IDDM.
Collapse
Affiliation(s)
- Jinhyuk Choi
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Fritz Cayabyab
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harvey Perez
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
16
|
Ding Y, Liu S, Liu J, Jin S, Wang J. Cryopreservation with DMSO affects the DNA integrity, apoptosis, cell cycle and function of human bone mesenchymal stem cells. Cryobiology 2024; 114:104847. [PMID: 38246511 DOI: 10.1016/j.cryobiol.2024.104847] [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: 05/11/2023] [Revised: 09/26/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
Cryopreservation (CP) enables pooling and long-term banking of various types of cells, which is indispensable for the cell therapeutics. Dimethyl sulfoxide (DMSO) is universally used as a cryoprotectant in basic and clinical research. Although, the use of DMSO has been under serious debate due to significant clinical side effects correlated with infusions of cellular therapy products containing DMSO, the effect of CP with DMSO on the cell properties and functions remains unknown. Here, we experimentally found that the CP of human bone mesenchymal stem cells (hBMSCs) with 10 % DMSO results 10-15 % of cells apoptosis upon immediate freeze-thaw, ca. 3.8 times of DNA damage/repair relative to the fresh ones after post-thaw cultured in 48 h, and cell cycle arrests at G0/G1 after post-thaw cultured in 24 h. Moreover, CP with 10 % DMSO significantly increases the reactive oxygen species (ROS) level of the frozen-thawed MSCs which may be one of the causes impair cellular properties and functions. Indeed, we found that the differentiation and migration ability of post-thaw cultured hBMSCs decrease as the expression of adipogenic, osteogenic genes and F-actin reduces in the comparison with those of the fresh cells.
Collapse
Affiliation(s)
- Yanqin Ding
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shuo Liu
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jianting Liu
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shenglin Jin
- Interdisciplinary Research Center, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100190, China; Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, China.
| | - Jianjun Wang
- Interdisciplinary Research Center, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100190, China; Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, China.
| |
Collapse
|
17
|
Sethia N, Rao JS, Khashim Z, Schornack AMR, Etheridge ML, Peterson QP, Finger EB, Bischof JC, Dutcher CS. On Chip Sorting of Stem Cell-Derived β Cell Clusters Using Traveling Surface Acoustic Waves. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40. [PMID: 38318799 PMCID: PMC10883307 DOI: 10.1021/acs.langmuir.3c02934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/05/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
There is a critical need for sorting complex materials, such as pancreatic islets of Langerhans, exocrine acinar tissues, and embryoid bodies. These materials are cell clusters, which have highly heterogeneous physical properties (such as size, shape, morphology, and deformability). Selecting such materials on the basis of specific properties can improve clinical outcomes and help advance biomedical research. In this work, we focused on sorting one such complex material, human stem cell-derived β cell clusters (SC-β cell clusters), by size. For this purpose, we developed a microfluidic device in which an image detection system was coupled to an actuation mechanism based on traveling surface acoustic waves (TSAWs). SC-β cell clusters of varying size (∼100-500 μm in diameter) were passed through the sorting device. Inside the device, the size of each cluster was estimated from their bright-field images. After size identification, larger clusters, relative to the cutoff size for separation, were selectively actuated using TSAW pulses. As a result of this selective actuation, smaller and larger clusters exited the device from different outlets. At the current sample dilutions, the experimental sorting efficiency ranged between 78% and 90% for a separation cutoff size of 250 μm, yielding sorting throughputs of up to 0.2 SC-β cell clusters/s using our proof-of-concept design. The biocompatibility of this sorting technique was also established, as no difference in SC-β cell cluster viability due to TSAW pulse usage was found. We conclude the proof-of-concept sorting work by discussing a few ways to optimize sorting of SC-β cell clusters for potentially higher sorting efficiency and throughput. This sorting technique can potentially help in achieving a better distribution of islets for clinical islet transplantation (a potential cure for type 1 diabetes). Additionally, the use of this technique for sorting islets can help in characterizing islet biophysical properties by size and selecting suitable islets for improved islet cryopreservation.
Collapse
Affiliation(s)
- Nikhil Sethia
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph Sushil Rao
- Division
of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Schulze
Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zenith Khashim
- Department
of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Anna Marie R. Schornack
- Department
of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Michael L. Etheridge
- Department
of Mechanical Engineering, University of
Minnesota, Minneapolis, Minnesota 55455, United States
| | - Quinn P. Peterson
- Department
of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Center for
Regenerative Biotherapeutics, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Erik B. Finger
- Division
of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - John C. Bischof
- Department
of Mechanical Engineering, University of
Minnesota, Minneapolis, Minnesota 55455, United States
- Department
of Biomedical Engineering, University of
Minnesota, Minneapolis, Minnesota 55455, United States
| | - Cari S. Dutcher
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department
of Mechanical Engineering, University of
Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
18
|
Han H, Zhan T, Guo N, Cui M, Xu Y. Cryopreservation of organoids: Strategies, innovation, and future prospects. Biotechnol J 2024; 19:e2300543. [PMID: 38403430 DOI: 10.1002/biot.202300543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 02/27/2024]
Abstract
Organoid technology has demonstrated unique advantages in multidisciplinary fields such as disease research, tumor drug sensitivity, clinical immunity, drug toxicology, and regenerative medicine. It will become the most promising research tool in translational research. However, the long preparation time of organoids and the lack of high-quality cryopreservation methods limit the further application of organoids. Although the high-quality cryopreservation of small-volume biological samples such as cells and embryos has been successfully achieved, the existing cryopreservation methods for organoids still face many bottlenecks. In recent years, with the development of materials science, cryobiology, and interdisciplinary research, many new materials and methods have been applied to cryopreservation. Several new cryopreservation methods have emerged, such as cryoprotectants (CPAs) of natural origin, ice-controlled biomaterials, and rapid rewarming methods. The introduction of these technologies has expanded the research scope of cryopreservation of organoids, provided new approaches and methods for cryopreservation of organoids, and is expected to break through the current technical bottleneck of cryopreservation of organoids. This paper reviews the progress of cryopreservation of organoids in recent years from three aspects: damage factors of cryopreservation of organoids, new protective agents and loading methods, and new technologies of cryopreservation and rewarming.
Collapse
Affiliation(s)
- Hengxin Han
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Taijie Zhan
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Ning Guo
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Mengdong Cui
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Yi Xu
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| |
Collapse
|
19
|
Guo Z, Zuchowicz N, Bouwmeester J, Joshi AS, Neisch AL, Smith K, Daly J, Etheridge ML, Finger EB, Kodandaramaiah SB, Hays TS, Hagedorn M, Bischof JC. Conduction-Dominated Cryomesh for Organism Vitrification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303317. [PMID: 38018294 PMCID: PMC10797434 DOI: 10.1002/advs.202303317] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/20/2023] [Indexed: 11/30/2023]
Abstract
Vitrification-based cryopreservation is a promising approach to achieving long-term storage of biological systems for maintaining biodiversity, healthcare, and sustainable food production. Using the "cryomesh" system achieves rapid cooling and rewarming of biomaterials, but further improvement in cooling rates is needed to increase biosystem viability and the ability to cryopreserve new biosystems. Improved cooling rates and viability are possible by enabling conductive cooling through cryomesh. Conduction-dominated cryomesh improves cooling rates from twofold to tenfold (i.e., 0.24 to 1.2 × 105 °C min-1 ) in a variety of biosystems. Higher thermal conductivity, smaller mesh wire diameter and pore size, and minimizing the nitrogen vapor barrier (e.g., vertical plunging in liquid nitrogen) are key parameters to achieving improved vitrification. Conduction-dominated cryomesh successfully vitrifies coral larvae, Drosophila embryos, and zebrafish embryos with improved outcomes. Not only a theoretical foundation for improved vitrification in µm to mm biosystems but also the capability to scale up for biorepositories and/or agricultural, aquaculture, or scientific use are demonstrated.
Collapse
Affiliation(s)
- Zongqi Guo
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - Nikolas Zuchowicz
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - Jessica Bouwmeester
- Hawaii Institute of Marine BiologyUniversity of HawaiiKaneoheHI96744USA
- Smithsonian National Zoo and Conservation Biology InstituteFront RoyalVA22630USA
| | - Amey S. Joshi
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - Amanda L. Neisch
- Department of GeneticsCell Biology and DevelopmentUniversity of MinnesotaMinneapolisMN55455USA
| | - Kieran Smith
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - Jonathan Daly
- Taronga Conservation Society AustraliaMosmanNew South Wales2088Australia
- School of BiologicalEarth and Environmental SciencesUniversity of New South WalesKensingtonNew South Wales2033Australia
| | - Michael L. Etheridge
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - Erik B. Finger
- Department of SurgeryUniversity of MinnesotaMinneapolisMN55455USA
| | - Suhasa B. Kodandaramaiah
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
- Department of Biomedical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
- Graduate Program in NeuroscienceUniversity of MinnesotaMinneapolisMN55455USA
| | - Thomas S. Hays
- Department of GeneticsCell Biology and DevelopmentUniversity of MinnesotaMinneapolisMN55455USA
| | - Mary Hagedorn
- Hawaii Institute of Marine BiologyUniversity of HawaiiKaneoheHI96744USA
- Smithsonian National Zoo and Conservation Biology InstituteFront RoyalVA22630USA
| | - John C. Bischof
- Department of Mechanical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
- Department of Biomedical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
- Institute for Engineering in MedicineUniversity of MinnesotaMinneapolisMN55455USA
| |
Collapse
|
20
|
Czarnecka Z, Dadheech N, Razavy H, Pawlick R, Shapiro AMJ. The Current Status of Allogenic Islet Cell Transplantation. Cells 2023; 12:2423. [PMID: 37887267 PMCID: PMC10605704 DOI: 10.3390/cells12202423] [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: 09/12/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
Type 1 Diabetes (T1D) is an autoimmune destruction of pancreatic beta cells. The development of the Edmonton Protocol for islet transplantation in 2000 revolutionized T1D treatment and offered a glimpse at a cure for the disease. In 2022, the 20-year follow-up findings of islet cell transplantation demonstrated the long-term safety of islet cell transplantation despite chronic immunosuppression. The Edmonton Protocol, however, remains limited by two obstacles: scarce organ donor availability and risks associated with chronic immunosuppression. To overcome these challenges, the search has begun for an alternative cell source. In 2006, pluripotency genomic factors, coined "Yamanaka Factors," were discovered, which reprogram mature somatic cells back to their embryonic, pluripotent form (iPSC). iPSCs can then be differentiated into specialized cell types, including islet cells. This discovery has opened a gateway to a personalized medicine approach to treating diabetes, circumventing the issues of donor supply and immunosuppression. In this review, we present a brief history of allogenic islet cell transplantation from the early days of pancreatic remnant transplantation to present work on encapsulating stem cell-derived cells. We review data on long-term outcomes and the ongoing challenges of allogenic islet cell and stem cell-derived islet cell transplant.
Collapse
Affiliation(s)
- Zofia Czarnecka
- Department of Surgery, University of Alberta, Edmonton, AB T6G 2RW3, Canada; (N.D.); (H.R.); (R.P.); (A.M.J.S.)
| | | | | | | | | |
Collapse
|
21
|
Weng L. Cell Therapy Drug Product Development: Technical Considerations and Challenges. J Pharm Sci 2023; 112:2615-2620. [PMID: 37549846 DOI: 10.1016/j.xphs.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
Cell therapy uses living cells as a drug to treat diseases. To develop a cell therapy drug product (DP), cryopreservation plays a central role in extending the shelf life of these living medicines by pausing their biological activities, especially preventing degradation, at a temperature as low as liquid nitrogen. This helps overcome the temporal and geographical gaps between centralized manufacturing and clinical administration, as well as allowing sufficient time for full release testing and flexibility in scheduling patients for administration. Cryopreservation determines or influences several key manufacturing, logistical, or clinical in-use processes, including formulation, filling, controlled rate freezing, cryogenic storage and transportation, thawing, and dose preparation. This article overviews the key technical aspects of cell therapy DP development and elucidates fundamental principles of cryobiology that should be considered when we design and optimize the relevant processes. This article also discusses the challenges that motivate continued innovation for cell therapy drug product development.
Collapse
Affiliation(s)
- Lindong Weng
- Novo Nordisk Research Center Seattle, Inc., United States.
| |
Collapse
|
22
|
Han Z, Rao JS, Ramesh S, Hergesell J, Namsrai BE, Etheridge ML, Finger EB, Bischof JC. Model-Guided Design and Optimization of CPA Perfusion Protocols for Whole Organ Cryopreservation. Ann Biomed Eng 2023; 51:2216-2228. [PMID: 37351756 PMCID: PMC10518287 DOI: 10.1007/s10439-023-03255-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023]
Abstract
Vitrification could enable long-term organ preservation, but only after loading high-concentration, potentially toxic cryoprotective agents (CPAs) by perfusion. In this paper, we combine a two-compartment Krogh cylinder model with a toxicity cost function to theoretically optimize the loading of CPA (VMP) in rat kidneys as a model system. First, based on kidney perfusion experiments, we systematically derived the parameters for a CPA transport loading model, including the following: Vb = 86.0% (ra = 3.86 μm), Lp = 1.5 × 10-14 m3/(N·s), ω = 7.0 × 10-13 mol/(N·s), σ = 0.10. Next, we measured the toxicity cost function model parameters as α = 3.12 and β = 9.39 × 10-6. Combining these models, we developed an improved kidney-loading protocol predicted to achieve vitrification while minimizing toxicity. The optimized protocol resulted in shorter exposure (25 min or 18.5% less) than the gold standard kidney-loading protocol for VMP, which had been developed based on decades of empirical practice. After testing both protocols on rat kidneys, we found comparable physical and biological outcomes. While we did not dramatically reduce toxicity, we did reduce the time. As our approach is now validated, it can be used on other organs lacking defined toxicity data to reduce CPA exposure time and provide a rapid path toward developing CPA perfusion protocols for other organs and CPAs.
Collapse
Affiliation(s)
- Zonghu Han
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Joseph Sushil Rao
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, USA
| | - Srivasupradha Ramesh
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Jan Hergesell
- Institute for Multiphase Processes (IMP), Leibniz University, Hannover, Germany
| | | | - Michael L Etheridge
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Erik B Finger
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA.
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, USA.
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
23
|
Powell-Palm MJ, Henley EM, Consiglio AN, Lager C, Chang B, Perry R, Fitzgerald K, Daly J, Rubinsky B, Hagedorn M. Cryopreservation and revival of Hawaiian stony corals using isochoric vitrification. Nat Commun 2023; 14:4859. [PMID: 37612315 PMCID: PMC10447501 DOI: 10.1038/s41467-023-40500-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Abstract
Corals are under siege by both local and global threats, creating a worldwide reef crisis. Cryopreservation is an important intervention measure and a vital component of the modern coral conservation toolkit, but preservation techniques are currently limited to sensitive reproductive materials that can only be obtained a few nights per year during spawning. Here, we report the successful cryopreservation and revival of cm-scale coral fragments via mL-scale isochoric vitrification. We demonstrate coral viability at 24 h post-thaw using a calibrated oxygen-uptake respirometry technique, and further show that the method can be applied in a passive, electronics-free configuration. Finally, we detail a complete prototype coral cryopreservation pipeline, which provides a platform for essential next steps in modulating post-thaw stress and initiating long-term growth. These findings pave the way towards an approach that can be rapidly deployed around the world to secure the biological genetic diversity of our vanishing coral reefs.
Collapse
Affiliation(s)
- Matthew J Powell-Palm
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX, USA.
| | - E Michael Henley
- Smithsonian National Zoo and Conservation Biology Institute, Front Royal, VA, 22630, USA.
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, 96744, USA.
| | - Anthony N Consiglio
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Claire Lager
- Smithsonian National Zoo and Conservation Biology Institute, Front Royal, VA, 22630, USA
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, 96744, USA
| | - Brooke Chang
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Riley Perry
- Smithsonian National Zoo and Conservation Biology Institute, Front Royal, VA, 22630, USA
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, 96744, USA
| | - Kendall Fitzgerald
- Smithsonian National Zoo and Conservation Biology Institute, Front Royal, VA, 22630, USA
| | - Jonathan Daly
- Taronga Institute of Science and Learning, Taronga Conservation Society Australia, Mosman, NSW, 2088, Australia
- Centre for Ecosystem Science and Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Boris Rubinsky
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Mary Hagedorn
- Smithsonian National Zoo and Conservation Biology Institute, Front Royal, VA, 22630, USA
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, 96744, USA
| |
Collapse
|
24
|
Zhan T, Niu W, Cui M, Han H, Dang H, Guo N, Wang D, Hao Y, Zang C, Xu Y, Guo H. A study on the relationship between the crystallization characteristics of quenched droplets and the effect of cell cryopreservation with Raman spectroscopy. Analyst 2023. [PMID: 37337775 DOI: 10.1039/d3an00652b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
The cryopreservation method of microdroplets has steadily become widely employed in the cryopreservation of microscale biological samples such as various types of cells due to its fast cooling rate, significant reduction of the concentration of cryoprotectants, and practical liquid handling method. However, it is still necessary to consider the corresponding relationship between droplet size and concentration and the impact of crystallization during the cooling process on cell viability. The key may be a misunderstanding of the influencing factors of crystallization and vitrification behavior with concentration during cooling on the ultimate cell viability, which may be attributable to the inability to analyze the freezing state inside the microdroplets. Therefore, in this work, an in situ Raman observation system for droplet quenching was assembled to obtain Raman spectra in the frozen state, and the spectral characteristics of the crystallization and vitrification processes of microdroplets with varied concentrations and volumes were investigated. Furthermore, the degree of crystallization inside the droplets was quantitatively analyzed, and it was found that the ratio of the crystalline peak to hydrogen bond shoulder could clearly distinguish the degree of crystallization and the vitrified state, and the Raman crystallization characteristic parameters gradually increased with the decrease of concentrations. By obtaining the cooling curve and the overall cooling rate of quenching droplets, the vitrification state of the microdroplets was confirmed by theoretical analysis of the cooling characteristics of a DMSO solution system. In addition, the effect of cell cryopreservation was investigated using the microdroplet quenching device, and it was found that the key to cell survival during the quenching process of low-concentration microdroplets was dominated by the cooling rate and the internal crystallization degree, while the main influencing factor on high concentration was the toxic effect of a protective agent. In general, this work introduces a new nondestructive evaluation and analysis method for the cryopreservation of quenching microdroplets.
Collapse
Affiliation(s)
- Taijie Zhan
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Wenya Niu
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Mengdong Cui
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Hengxin Han
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Hangyu Dang
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Ning Guo
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Ding Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yan Hao
- Yinfeng Cryomedicine Technology Co. Ltd, Jinan, China
| | - Chuanbao Zang
- Yinfeng Cryomedicine Technology Co. Ltd, Jinan, China
| | - Yi Xu
- Institute of Bio-thermal Science and Technology, Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai Technical Service Platform for Cryopreservation of Biological Resources, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Hanming Guo
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
25
|
Zhang W, Liu X, Hu Y, Tan S. Incorporate delivery, warming and washing methods into efficient cryopreservation. Front Bioeng Biotechnol 2023; 11:1215591. [PMID: 37397963 PMCID: PMC10309563 DOI: 10.3389/fbioe.2023.1215591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/08/2023] [Indexed: 07/04/2023] Open
|
26
|
Chen J, Liu X, Hu Y, Chen X, Tan S. Cryopreservation of tissues and organs: present, bottlenecks, and future. Front Vet Sci 2023; 10:1201794. [PMID: 37303729 PMCID: PMC10248239 DOI: 10.3389/fvets.2023.1201794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Tissue and organ transplantation continues to be an effective measure for saving the lives of certain critically ill patients. The organ preservation methods that are commonly utilized in clinical practice are presently only capable of achieving short-term storage, which is insufficient for meeting the demand for organ transplantation. Ultra-low temperature storage techniques have garnered significant attention due to their capacity for achieving long-term, high-quality preservation of tissues and organs. However, the experience of cryopreserving cells cannot be readily extrapolated to the cryopreservation of complex tissues and organs, and the latter still confronts numerous challenges in its clinical application. This article summarizes the current research progress in the cryogenic preservation of tissues and organs, discusses the limitations of existing studies and the main obstacles facing the cryopreservation of complex tissues and organs, and finally introduces potential directions for future research efforts.
Collapse
|
27
|
Hogrebe NJ, Ishahak M, Millman JR. Developments in stem cell-derived islet replacement therapy for treating type 1 diabetes. Cell Stem Cell 2023; 30:530-548. [PMID: 37146579 PMCID: PMC10167558 DOI: 10.1016/j.stem.2023.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
The generation of islet-like endocrine clusters from human pluripotent stem cells (hPSCs) has the potential to provide an unlimited source of insulin-producing β cells for the treatment of diabetes. In order for this cell therapy to become widely adopted, highly functional and well-characterized stem cell-derived islets (SC-islets) need to be manufactured at scale. Furthermore, successful SC-islet replacement strategies should prevent significant cell loss immediately following transplantation and avoid long-term immune rejection. This review highlights the most recent advances in the generation and characterization of highly functional SC-islets as well as strategies to ensure graft viability and safety after transplantation.
Collapse
Affiliation(s)
- Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA.
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA.
| |
Collapse
|
28
|
Sharma A, Lee CY, Namsrai BE, Han Z, Tobolt D, Rao JS, Gao Z, Etheridge ML, Garwood M, Clemens MG, Bischof JC, Finger EB. Cryopreservation of Whole Rat Livers by Vitrification and Nanowarming. Ann Biomed Eng 2023; 51:566-577. [PMID: 36183025 PMCID: PMC10315167 DOI: 10.1007/s10439-022-03064-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/22/2022] [Indexed: 11/01/2022]
Abstract
Liver cryopreservation has the potential to enable indefinite organ banking. This study investigated vitrification-the ice-free cryopreservation of livers in a glass-like state-as a promising alternative to conventional cryopreservation, which uniformly fails due to damage from ice formation or cracking. Our unique "nanowarming" technology, which involves perfusing biospecimens with cryoprotective agents (CPAs) and silica-coated iron oxide nanoparticles (sIONPs) and then, after vitrification, exciting the nanoparticles via radiofrequency waves, enables rewarming of vitrified specimens fast enough to avoid ice formation and uniformly enough to prevent cracking from thermal stresses, thereby addressing the two main failures of conventional cryopreservation. This study demonstrates the ability to load rat livers with both CPA and sIONPs by vascular perfusion, cool them rapidly to an ice-free vitrified state, and rapidly and homogenously rewarm them. While there was some elevation of liver enzymes (Alanine Aminotransferase) and impaired indocyanine green (ICG) excretion, the nanowarmed livers were viable, maintained normal tissue architecture, had preserved vascular endothelium, and demonstrated hepatocyte and organ-level function, including production of bile and hepatocyte uptake of ICG during normothermic reperfusion. These findings suggest that cryopreservation of whole livers via vitrification and nanowarming has the potential to achieve organ banking for transplant and other biomedical applications.
Collapse
Affiliation(s)
- Anirudh Sharma
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Charles Y Lee
- Department of Mechanical Engineering and Engineering Science, University of North Carolina, Charlotte, NC, 28223, USA
- Center for Biomedical Engineering and Science, University of North Carolina, Charlotte, NC, 28223, USA
| | - Bat-Erdene Namsrai
- Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Zonghu Han
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Diane Tobolt
- Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joseph Sushil Rao
- Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
- Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Zhe Gao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Michael L Etheridge
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Michael Garwood
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Mark G Clemens
- Center for Biomedical Engineering and Science, University of North Carolina, Charlotte, NC, 28223, USA
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, 28223, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Erik B Finger
- Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA.
- Division of Solid Organ Transplantation, University of Minnesota, 420 Delaware St. S.E., MMC 195, Minneapolis, MN, 55455, USA.
| |
Collapse
|
29
|
Rao JS, Pruett TL. Immunology of the transplanted cryopreserved kidney. Cryobiology 2023; 110:1-7. [PMID: 36640932 DOI: 10.1016/j.cryobiol.2023.01.003] [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: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Transplantation has substituted dysfunctional organs with healthy organs from donors to significantly lower morbidity and mortality associated with end-stage organ disease. Since the advent of transplantation, the promise of functional replacement has attracted an exponential mismatch between organ supply and demand. Theoretical proposals to counter the increasing needs have either been to create a source through genetic engineering of porcine donors for xenotransplantation (with more potent immunosuppression protocols) or recreate one's organ in a pig using interspecies blastocyst complementation for exogenic organ transplantation (without immunosuppression). Another promising avenue has been organ banking through cryopreservation for transplantation. Although ice free preservation and acceptable early function following rewarming is critical for success in transplantation, the immunological response that predominantly defines short- and long-term graft survival has failed to captivate attention to date. It is well sorted that thermal and metabolic stress incurred at 4 °C during recovery and reperfusion of organs for clinical transplantation has varying impact on graft survival. Considering the magnitude of cellular imbalance and injury at sub-zero/ultralow temperatures in addition to the chemical toxicity of cryoprotective agents (CPA), it is essential to assess and address the immunological response associated following transplantation to maximize the success of cryopreservation.
Collapse
Affiliation(s)
- Joseph Sushil Rao
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | - Timothy L Pruett
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
30
|
Marchini A, Ciulla MG, Antonioli B, Agnoli A, Bovio U, Visnoviz V, Bertuzzi F, Gelain F. Long-term cultures of human pancreatic islets in self-assembling peptides hydrogels. Front Bioeng Biotechnol 2023; 11:1105157. [PMID: 36911193 PMCID: PMC9995881 DOI: 10.3389/fbioe.2023.1105157] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Human pancreatic islets transplantation is an experimental therapeutic treatment for Type I Diabetes. Limited islets lifespan in culture remains the main drawback, due to the absence of native extracellular matrix as mechanical support after their enzymatic and mechanical isolation procedure. Extending the limited islets lifespan by creating a long-term in vitro culture remains a challenge. In this study, three biomimetic self-assembling peptides were proposed as potential candidates to recreate in vitro a pancreatic extracellular matrix, with the aim to mechanically and biologically support human pancreatic islets, by creating a three-dimensional culture system. The embedded human islets were analyzed for morphology and functionality in long-term cultures (14-and 28-days), by evaluating β-cells content, endocrine component, and extracellular matrix constituents. The three-dimensional support provided by HYDROSAP scaffold, and cultured into MIAMI medium, displayed a preserved islets functionality, a maintained rounded islets morphology and an invariable islets diameter up to 4 weeks, with results analogues to freshly-isolated islets. In vivo efficacy studies of the in vitro 3D cell culture system are ongoing; however, preliminary data suggest that human pancreatic islets pre-cultured for 2 weeks in HYDROSAP hydrogels and transplanted under subrenal capsule may restore normoglycemia in diabetic mice. Therefore, engineered self-assembling peptide scaffolds may provide a useful platform for long-term maintenance and preservation of functional human pancreatic islets in vitro.
Collapse
Affiliation(s)
- Amanda Marchini
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Maria Gessica Ciulla
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Barbara Antonioli
- Tissue Bank and Tissue Therapy Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessandro Agnoli
- Department of Biotechnology and Bioscience, University of Milan-Bicocca, Milan, Italy
| | - Umberto Bovio
- Department of Biotechnology and Bioscience, University of Milan-Bicocca, Milan, Italy
| | | | - Federico Bertuzzi
- Department of Diabetology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Fabrizio Gelain
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
31
|
Halliez C, Ibrahim H, Otonkoski T, Mallone R. In vitro beta-cell killing models using immune cells and human pluripotent stem cell-derived islets: Challenges and opportunities. Front Endocrinol (Lausanne) 2023; 13:1076683. [PMID: 36726462 PMCID: PMC9885197 DOI: 10.3389/fendo.2022.1076683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Type 1 diabetes (T1D) is a disease of both autoimmunity and β-cells. The β-cells play an active role in their own demise by mounting defense mechanisms that are insufficient at best, and that can become even deleterious in the long term. This complex crosstalk is important to understanding the physiological defense mechanisms at play in healthy conditions, their alterations in the T1D setting, and therapeutic agents that may boost such mechanisms. Robust protocols to develop stem-cell-derived islets (SC-islets) from human pluripotent stem cells (hPSCs), and islet-reactive cytotoxic CD8+ T-cells from peripheral blood mononuclear cells offer unprecedented opportunities to study this crosstalk. Challenges to develop in vitro β-cell killing models include the cluster morphology of SC-islets, the relatively weak cytotoxicity of most autoimmune T-cells and the variable behavior of in vitro expanded CD8+ T-cells. These challenges may however be highly rewarding in light of the opportunities offered by such models. Herein, we discuss these opportunities including: the β-cell/immune crosstalk in an islet microenvironment; the features that make β-cells more sensitive to autoimmunity; therapeutic agents that may modulate β-cell vulnerability; and the possibility to perform analyses in an autologous setting, i.e., by generating T-cell effectors and SC-islets from the same donor.
Collapse
Affiliation(s)
- Clémentine Halliez
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
- Department of Pediatrics, Helsinki University Hospital, Helsinki, Finland
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| |
Collapse
|
32
|
Engineering Strategies of Islet Product for Endocrine Regeneration. ENGINEERED REGENERATION 2023. [DOI: 10.1016/j.engreg.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
33
|
Lin M, Cao H, Li J. Control strategies of ice nucleation, growth, and recrystallization for cryopreservation. Acta Biomater 2023; 155:35-56. [PMID: 36323355 DOI: 10.1016/j.actbio.2022.10.056] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 02/02/2023]
Abstract
The cryopreservation of biomaterials is fundamental to modern biotechnology and biomedicine, but the biggest challenge is the formation of ice, resulting in fatal cryoinjury to biomaterials. To date, abundant ice control strategies have been utilized to inhibit ice formation and thus improve cryopreservation efficiency. This review focuses on the mechanisms of existing control strategies regulating ice formation and the corresponding applications to biomaterial cryopreservation, which are of guiding significance for the development of ice control strategies. Herein, basics related to biomaterial cryopreservation are introduced first. Then, the theoretical bases of ice nucleation, growth, and recrystallization are presented, from which the key factors affecting each process are analyzed, respectively. Ice nucleation is mainly affected by melting temperature, interfacial tension, shape factor, and kinetic prefactor, and ice growth is mainly affected by solution viscosity and cooling/warming rate, while ice recrystallization is inhibited by adsorption or diffusion mechanisms. Furthermore, the corresponding research methods and specific control strategies for each process are summarized. The review ends with an outlook of the current challenges and future perspectives in cryopreservation. STATEMENT OF SIGNIFICANCE: Ice formation is the major limitation of cryopreservation, which causes fatal cryoinjury to cryopreserved biomaterials. This review focuses on the three processes related to ice formation, called nucleation, growth, and recrystallization. The theoretical models, key influencing factors, research methods and corresponding ice control strategies of each process are summarized and discussed, respectively. The systematic introduction on mechanisms and control strategies of ice formation is instructive for the cryopreservation development.
Collapse
Affiliation(s)
- Min Lin
- Key Laboratory for Thermal Science and Power Engineering of Ministry of Education, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for CO(2) Utilization and Reduction Technology, Tsinghua University, Beijing 100084, China
| | - Haishan Cao
- Key Laboratory for Thermal Science and Power Engineering of Ministry of Education, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for CO(2) Utilization and Reduction Technology, Tsinghua University, Beijing 100084, China.
| | - Junming Li
- Key Laboratory for Thermal Science and Power Engineering of Ministry of Education, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for CO(2) Utilization and Reduction Technology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
34
|
Xu R, Treeby BE, Martin E. Experiments and simulations demonstrating the rapid ultrasonic rewarming of frozen tissue cryovials. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2023; 153:517. [PMID: 36732249 DOI: 10.1121/10.0016886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/02/2023] [Indexed: 06/18/2023]
Abstract
The development of methods to safely rewarm large cryopreserved biological samples remains a barrier to the widespread adoption of cryopreservation. Here, experiments and simulations were performed to demonstrate that ultrasound can increase rewarming rates relative to thermal conduction alone. An ultrasonic rewarming setup based on a custom 444 kHz tubular piezoelectric transducer was designed, characterized, and tested with 2 ml cryovials filled with frozen ground beef. Rewarming rates were characterized in the -20 °C to 5 °C range. Thermal conduction-based rewarming was compared to thermal conduction plus ultrasonic rewarming, demonstrating a tenfold increase in rewarming rate when ultrasound was applied. The maximum recorded rewarming rate with ultrasound was 57° C/min, approximately 2.5 times faster than with thermal conduction alone. Coupled acoustic and thermal simulations were developed and showed good agreement with the heating rates demonstrated experimentally and were also used to demonstrate spatial heating distributions with small (<3° C) temperature differentials throughout the sample when the sample was below 0° C. The experiments and simulations demonstrate the potential for ultrasonic cryovial rewarming with a possible application to large volume rewarming, as faster rewarming rates may improve the viability of cryopreserved tissues and reduce the time needed for cells to regain normal function.
Collapse
Affiliation(s)
- Rui Xu
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Bradley E Treeby
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Eleanor Martin
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| |
Collapse
|
35
|
Kuang G, Zhang Q, Jia J, Yu Y. Freezing biological organisms for biomedical applications. SMART MEDICINE 2022; 1:e20220034. [PMID: 39188743 PMCID: PMC11235656 DOI: 10.1002/smmd.20220034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 08/28/2024]
Abstract
Biological organisms play important roles in human health, either in a commensal or pathogenic manner. Harnessing inactivated organisms or living organisms is a promising way to treat diseases. As two types of freezing, cryoablation makes it simple to inactivate organisms that must be in a non-pathogenic state when needed, while cryopreservation is a facile way to address the problem of long-term storage challenged by living organism-based therapy. In this review, we present the latest studies of freezing biological organisms for biomedical applications. To begin with, the freezing strategies of cryoablation and cryopreservation, as well as their corresponding technical essentials, are illustrated. Besides, biomedical applications of freezing biological organisms are presented, including transplantation, tissue regeneration, anti-infection therapy, and anti-tumor therapy. The challenges and prospects of freezing living organisms for biomedical applications are well discussed. We believe that the freezing method will provide a potential direction for the standardization and commercialization of inactivated or living organism-based therapeutic systems, and promote the clinical application of organism-based therapy.
Collapse
Affiliation(s)
- Gaizhen Kuang
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Qingfei Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Jinxuan Jia
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Yunru Yu
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
| |
Collapse
|
36
|
Leavens KF, Alvarez-Dominguez JR, Vo LT, Russ HA, Parent AV. Stem cell-based multi-tissue platforms to model human autoimmune diabetes. Mol Metab 2022; 66:101610. [PMID: 36209784 PMCID: PMC9587366 DOI: 10.1016/j.molmet.2022.101610] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/20/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic insulin-producing β cells are specifically destroyed by the immune system. Understanding the initiation and progression of human T1D has been hampered by the lack of appropriate models that can reproduce the complexity and heterogeneity of the disease. The development of platforms combining multiple human pluripotent stem cell (hPSC) derived tissues to model distinct aspects of T1D has the potential to provide critical novel insights into the etiology and pathogenesis of the human disease. SCOPE OF REVIEW In this review, we summarize the state of hPSC differentiation approaches to generate cell types and tissues relevant to T1D, with a particular focus on pancreatic islet cells, T cells, and thymic epithelium. We present current applications as well as limitations of using these hPSC-derived cells for disease modeling and discuss efforts to optimize platforms combining multiple cell types to model human T1D. Finally, we outline remaining challenges and emphasize future improvements needed to accelerate progress in this emerging field of research. MAJOR CONCLUSIONS Recent advances in reprogramming approaches to create patient-specific induced pluripotent stem cell lines (iPSCs), genome engineering technologies to efficiently modify DNA of hPSCs, and protocols to direct their differentiation into mature cell types have empowered the use of stem cell derivatives to accurately model human disease. While challenges remain before complex interactions occurring in human T1D can be modeled with these derivatives, experiments combining hPSC-derived β cells and immune cells are already providing exciting insight into how these cells interact in the context of T1D, supporting the viability of this approach.
Collapse
Affiliation(s)
- Karla F Leavens
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania and Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Juan R Alvarez-Dominguez
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Linda T Vo
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Audrey V Parent
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
37
|
Biomolecular Pathways of Cryoinjuries in Low-Temperature Storage for Mammalian Specimens. Bioengineering (Basel) 2022; 9:bioengineering9100545. [PMID: 36290513 PMCID: PMC9598205 DOI: 10.3390/bioengineering9100545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
Low-temperature preservation could effectively extend in vitro storage of biological materials due to delayed or suspended cellular metabolism and decaying as illustrated by the Arrhenius model. It is widely used as an enabling technology for a variety of biomedical applications such as cell therapeutics, assisted reproductive technologies, organ transplantation, and mRNA medicine. Although the technology to minimize cryoinjuries of mammalian specimens during preservation has been advanced substantially over past decades, mammalian specimens still suffer cryoinjuries under low-temperature conditions. Particularly, the molecular mechanisms underlying cryoinjuries are still evasive, hindering further improvement and development of preservation technologies. In this paper, we systematically recapitulate the molecular cascades of cellular injuries induced by cryopreservation, including apoptosis, necroptosis, ischemia-reperfusion injury (IRI). Therefore, this study not only summarizes the impact of low-temperature preservations on preserved cells and organs on the molecular level, but also provides a molecular basis to reduce cryoinjuries for future exploration of biopreservation methods, materials, and devices.
Collapse
|
38
|
Zhan L, Han Z, Shao Q, Etheridge ML, Hays T, Bischof JC. Rapid joule heating improves vitrification based cryopreservation. Nat Commun 2022; 13:6017. [PMID: 36224179 PMCID: PMC9556611 DOI: 10.1038/s41467-022-33546-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/21/2022] [Indexed: 01/24/2023] Open
Abstract
Cryopreservation by vitrification has far-reaching implications. However, rewarming techniques that are rapid and scalable (both in throughput and biosystem size) for low concentrations of cryoprotective agent (CPA) for reduced toxicity are lacking, limiting the potential for translation. Here, we introduce a joule heating-based platform technology, whereby biosystems are rapidly rewarmed by contact with an electrical conductor that is fed a voltage pulse. We demonstrate successful cryopreservation of three model biosystems with thicknesses across three orders of magnitude, including adherent cells (~4 µm), Drosophila melanogaster embryos (~50 µm) and rat kidney slices (~1.2 mm) using low CPA concentrations (2-4 M). Using tunable voltage pulse widths from 10 µs to 100 ms, numerical simulation predicts that warming rates from 5 × 104 to 6 × 108 °C/min can be achieved. Altogether, our results present a general solution to the cryopreservation of a broad spectrum of cellular, organismal and tissue-based biosystems.
Collapse
Affiliation(s)
- Li Zhan
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA.
- Center for Engineering in Medicine, Massachusetts General Hospital, Shriners Hospital for Children, Harvard Medical School, Boston, MA, USA.
| | - Zonghu Han
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Qi Shao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Michael L Etheridge
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Thomas Hays
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA.
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
39
|
Wakabayashi T, Kaneko M, Nakai T, Horie M, Fujimoto H, Takahashi M, Tanoue S, Ito A. Nanowarming of vitrified pancreatic islets as a cryopreservation technology for transplantation. Bioeng Transl Med 2022. [DOI: 10.1002/btm2.10416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Taisei Wakabayashi
- Department of Chemical Systems Engineering, School of Engineering Nagoya University Nagoya Japan
| | - Masahiro Kaneko
- Department of Chemical Systems Engineering, School of Engineering Nagoya University Nagoya Japan
| | - Tomoki Nakai
- Department of Chemical Systems Engineering, School of Engineering Nagoya University Nagoya Japan
| | - Masanobu Horie
- Radioisotope Research Center, Agency of Health, Safety and Environment Kyoto University Kyoto Japan
| | - Hiroyuki Fujimoto
- Radioisotope Research Center, Agency of Health, Safety and Environment Kyoto University Kyoto Japan
| | | | - Shota Tanoue
- Technical Department Dai‐Ichi High Frequency Co., Ltd Kawasaki Japan
| | - Akira Ito
- Department of Chemical Systems Engineering, School of Engineering Nagoya University Nagoya Japan
| |
Collapse
|
40
|
Research Highlights. Transplantation 2022; 106:1711-1712. [PMID: 36735266 DOI: 10.1097/tp.0000000000004308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
41
|
Hu Q, Wang Z, Shen L, Zhao G. Label-Free and Noninvasive Single-Cell Characterization for the Viscoelastic Properties of Cryopreserved Human Red Blood Cells Using a Dielectrophoresis-On-a-Chip Approach. Anal Chem 2022; 94:10245-10255. [DOI: 10.1021/acs.analchem.2c01858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Qianqian Hu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei 230027, China
| | - Zirui Wang
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei 230027, China
| | - Lingxiao Shen
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei 230027, China
| | - Gang Zhao
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
42
|
Bai G, Hu J, Qin S, Qi Z, Zhuang H, Sun F, Lu Y, Jin S, Gao D, Wang J. Small-molecule fulvic acid with strong hydration ability for non-vitreous cellular cryopreservation. iScience 2022; 25:104423. [PMID: 35663038 PMCID: PMC9157229 DOI: 10.1016/j.isci.2022.104423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/12/2022] [Accepted: 05/11/2022] [Indexed: 12/11/2022] Open
Abstract
The exploitation of biocompatible ice-control materials especially the small molecules for non-vitreous cryopreservation remains challenging. Here, we report a small molecule of fulvic acid (FA) with strong hydration ability, which enables non-vitreous cellular cryopreservation by reducing ice growth during freezing and reducing ice recrystallization/promoting ice melting during thawing. Without adding any other cryoprotectants, FA can enhance the recovery of sheep red blood cells (RBCs) by three times as compared with a commercial cryoprotectant (hydroxyethyl starch) under a stringent test condition. Investigation of water mobility reveals that the ice-control properties of FA can be ascribed to its strong bondage to water molecules. Furthermore, we found that FA can be absorbed by RBCs and mainly locates on membranes, suggesting the possible contribution of FA to cell protection through stabilizing membranes. This work bespeaks a bright future for small-molecule cryoprotectants in non-vitreous cryopreservation application. FA shows strong hydration ability FA reduces ice growth/recrystallization and promotes ice melting FA can be absorbed by RBCs and mainly locates on membranes FA enables non-vitreous cellular cryopreservation
Collapse
Affiliation(s)
- Guoying Bai
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300401, China.,Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jinhao Hu
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Sijia Qin
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Zipeng Qi
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Hening Zhuang
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Fude Sun
- Key Laboratory of Hebei Province for Molecular Biophysics Institute of Biophysics, Hebei University of Technology, Tianjin 300401, China
| | - Youhua Lu
- Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Shenglin Jin
- Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Dong Gao
- Key Laboratory of Hebei Province for Molecular Biophysics Institute of Biophysics, Hebei University of Technology, Tianjin 300401, China
| | - Jianjun Wang
- Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
43
|
Scheibner K, Lickert H. Awaking sleeping islets for a cure of diabetes. MED 2022; 3:279-280. [DOI: 10.1016/j.medj.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Cold treatment for insulin-making cells points to diabetes cure. Nature 2022. [PMID: 35304886 DOI: 10.1038/d41586-022-00736-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|