1
|
Hu W, Wang Y, Han J, Zhang W, Chen J, Li X, Wang L. Microfluidic organ-on-a-chip models for the gut-liver axis: from structural mimicry to functional insights. Biomater Sci 2025; 13:1624-1656. [PMID: 40019226 DOI: 10.1039/d4bm01273a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The gut-liver axis plays a crucial role in maintaining metabolic balance and overall human health. It orchestrates various processes, such as blood flow, nutrient transfer, metabolite processing, and immune cell communication between the two organs. Traditional methods, such as animal models and two-dimensional (2D) cell cultures, are insufficient in fully replicating the intricate functions of the gut-liver axis. The emergence of microfluidic technology has revolutionized this field, facilitating the development of organ-on-a-chip (OOC) systems. These systems are capable of mimicking the complex structures and dynamic environments of the gut and liver in vitro and incorporating sensors for real-time monitoring. In this article, we review the latest progress in gut-on-a-chip (GOC) and liver-on-a-chip (LOC) systems, as well as the integrated gut-liver-on-a-chip (GLOC) models. Our focus lies in the simulation of physiological parameters, three-dimensional (3D) structural mimicry, microbiome integration, and multicellular co-culture. All these aspects are essential for constructing accurate in vitro models of the gut and liver. Furthermore, we explore the current applications of OOC technology in the study of the gut and liver, including its use in disease modeling, toxicity testing, and drug screening. Finally, we discuss the challenges that remain and outline potential future directions for advancing GOC and LOC development in vitro.
Collapse
Affiliation(s)
- Wanlin Hu
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
2
|
Hu T, Zhou T, Goit RK, Tam KC, Chan YK, Lam WC, Lo ACY. Bioactive Glial-Derived Neurotrophic Factor from a Safe Injectable Collagen-Alginate Composite Gel Rescues Retinal Photoreceptors from Retinal Degeneration in Rabbits. Mar Drugs 2024; 22:394. [PMID: 39330275 PMCID: PMC11433152 DOI: 10.3390/md22090394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
The management of vision-threatening retinal diseases remains challenging due to the lack of an effective drug delivery system. Encapsulated cell therapy (ECT) offers a promising approach for the continuous delivery of therapeutic agents without the need for immunosuppressants. In this context, an injectable and terminable collagen-alginate composite (CAC) ECT gel, designed with a Tet-on pro-caspase-8 system, was developed as a safe intraocular drug delivery platform for the sustained release of glial-cell-line-derived neurotrophic factor (GDNF) to treat retinal degenerative diseases. This study examined the potential clinical application of the CAC ECT gel, focusing on its safety, performance, and termination through doxycycline (Dox) administration in the eyes of healthy New Zealand White rabbits, as well as its therapeutic efficacy in rabbits with sodium-iodate (SI)-induced retinal degeneration. The findings indicated that the CAC ECT gel can be safely implanted without harming the retina or lens, displaying resistance to degradation, facilitating cell attachment, and secreting bioactive GDNF. Furthermore, the GDNF levels could be modulated by the number of implants. Moreover, Dox administration was effective in terminating gel function without causing retinal damage. Notably, rabbits with retinal degeneration treated with the gels exhibited significant functional recovery in both a-wave and b-wave amplitudes and showed remarkable efficacy in reducing photoreceptor apoptosis. Given its biocompatibility, mechanical stability, controlled drug release, terminability, and therapeutic effectiveness, our CAC ECT gel presents a promising therapeutic strategy for various retinal diseases in a clinical setting, eliminating the need for immunosuppressants.
Collapse
Affiliation(s)
- Tingyu Hu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Ting Zhou
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Rajesh Kumar Goit
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
- Jules Stein Eye Institute, Los Angeles, CA 90095, USA
| | - Ka Cheung Tam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Yau Kei Chan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Wai-Ching Lam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| |
Collapse
|
3
|
Çakıcı GT, Kaya S, Doğan SY, Solak EK. Quercetin-loaded sodium alginate/collagen/h-boron nitride potential wound dressings prepared using the Box-Behnken experimental design. Biotechnol J 2024; 19:e2300147. [PMID: 37897145 DOI: 10.1002/biot.202300147] [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: 04/01/2023] [Revised: 08/17/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND/AIMS Natural and synthetic biocompatible polymers have received significant attention in the pharmaceutical industry due to their rapid and effective healing properties in the wound healing process. The aim of this study was to optimize the extraction of onions, the preparation of sodium alginate/collagen/hydrogen boron nitride (NaAlg/Col/h-BN) membranes using the Box-Behnken experimental design, and determine the optimal conditions for quercetin release. The study also aimed to investigate the antimicrobial and antioxidant activities of the prepared membranes and their therapeutic properties. METHODS AND RESULTS The prepared membranes were characterized by scanning electron microscopy (SEM), fourier transform infrared (FTIR), differential scanning calorimetry (DSC), and X-ray diffraction (XRD). Antimicrobial activities were tested against Gram-negative (Gr-) Escherichia coli ATCC 25922, Klebsiella pneumonia, Enterobacter aerogenes, Gram-positive (Gr+) Staphylococcus aureus ATCC 25923, and Candida albicans ATCC 10231 pathogens. In vitro release studies were conducted to examine the therapeutic properties of the prepared membranes. The optimum conditions for the extraction of onions and the preparation of NaAlg/Col/h-BN membranes were found to be EtOH = 75 mL, t = 2 h, T = 45°C, and NaAlg = 1.0 g, Col = 2.0 g, and h-BN = 6% wt, respectively. The prepared membranes exhibited serious antimicrobial properties against S. aureus and C. albicans. The membranes also promoted the controlled release of quercetin for 24 h in vitro, indicating their potential as a new approach in wound treatment. CONCLUSION The study concludes that quercetin-filled NaAlg/Col/h-BN membranes have promising therapeutic properties for wound healing. The membranes exhibited significant antimicrobial and antioxidant properties, and their controlled release of quercetin suggests their potential for use in wound healing applications.
Collapse
Affiliation(s)
- Gülşen Taşkın Çakıcı
- Department of Chemistry and Chemical Processing Technologies, Vocational School of Technical Sciences, Gazi University, Ankara, Turkey
| | - Seçil Kaya
- Department of Material and Material Processing Technologies, Vocational School of Technical Sciences, Gazi University, Ankara, Turkey
| | - Sema Yiyit Doğan
- Department of Chemistry and Chemical Processing Technologies, Vocational School of Technical Sciences, Gazi University, Ankara, Turkey
| | - Ebru Kondolot Solak
- Department of Chemistry and Chemical Processing Technologies, Vocational School of Technical Sciences, Gazi University, Ankara, Turkey
- Department of Advanced Technologies, Gazi University, Ankara, Turkey
| |
Collapse
|
4
|
Pham CHL, Zuo Y, Chen Y, Tran NM, Nguyen DT, Dang TT. Waffle-inspired hydrogel-based macrodevice for spatially controlled distribution of encapsulated therapeutic microtissues and pro-angiogenic endothelial cells. Bioeng Transl Med 2023; 8:e10495. [PMID: 37206238 PMCID: PMC10189477 DOI: 10.1002/btm2.10495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/26/2022] [Accepted: 01/13/2023] [Indexed: 03/17/2023] Open
Abstract
Macro-encapsulation systems for delivery of cellular therapeutics in diabetes treatment offer major advantages such as device retrievability and high cell packing density. However, microtissue aggregation and absence of vasculature have been implicated in the inadequate transfer of nutrients and oxygen to the transplanted cellular grafts. Herein, we develop a hydrogel-based macrodevice to encapsulate therapeutic microtissues positioned in homogeneous spatial distribution to mitigate their aggregation while concurrently supporting an organized intra-device network of vascular-inductive cells. Termed Waffle-inspired Interlocking Macro-encapsulation (WIM) device, this platform comprises two modules with complementary topography features that fit together in a lock-and-key configuration. The waffle-inspired grid-like micropattern of the "lock" component effectively entraps insulin-secreting microtissues in controlled locations while the interlocking design places them in a co-planar spatial arrangement with close proximity to vascular-inductive cells. The WIM device co-laden with INS-1E microtissues and human umbilical vascular endothelial cells (HUVECs) maintains desirable cellular viability in vitro with the encapsulated microtissues retaining their glucose-responsive insulin secretion while embedded HUVECs express pro-angiogenic markers. Furthermore, a subcutaneously implanted alginate-coated WIM device encapsulating primary rat islets achieves blood glucose control for 2 weeks in chemically induced diabetic mice. Overall, this macrodevice design lays foundation for a cell delivery platform, which has the potential to facilitate nutrients and oxygen transport to therapeutic grafts and thereby might lead to improved disease management outcome.
Collapse
Affiliation(s)
- Chi H. L. Pham
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Yicong Zuo
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Yang Chen
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Nam M. Tran
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Dang T. Nguyen
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| | - Tram T. Dang
- School of Chemical and Biomedical EngineeringNanyang Technological University (NTU)SingaporeSingapore
| |
Collapse
|
5
|
Griffin KH, Fok SW, Kent Leach J. Strategies to capitalize on cell spheroid therapeutic potential for tissue repair and disease modeling. NPJ Regen Med 2022; 7:70. [PMID: 36494368 PMCID: PMC9734656 DOI: 10.1038/s41536-022-00266-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Cell therapies offer a tailorable, personalized treatment for use in tissue engineering to address defects arising from trauma, inefficient wound repair, or congenital malformation. However, most cell therapies have achieved limited success to date. Typically injected in solution as monodispersed cells, transplanted cells exhibit rapid cell death or insufficient retention at the site, thereby limiting their intended effects to only a few days. Spheroids, which are dense, three-dimensional (3D) aggregates of cells, enhance the beneficial effects of cell therapies by increasing and prolonging cell-cell and cell-matrix signaling. The use of spheroids is currently under investigation for many cell types. Among cells under evaluation, spheroids formed of mesenchymal stromal cells (MSCs) are particularly promising. MSC spheroids not only exhibit increased cell survival and retained differentiation, but they also secrete a potent secretome that promotes angiogenesis, reduces inflammation, and attracts endogenous host cells to promote tissue regeneration and repair. However, the clinical translation of spheroids has lagged behind promising preclinical outcomes due to hurdles in their formation, instruction, and use that have yet to be overcome. This review will describe the current state of preclinical spheroid research and highlight two key examples of spheroid use in clinically relevant disease modeling. It will highlight techniques used to instruct the phenotype and function of spheroids, describe current limitations to their use, and offer suggestions for the effective translation of cell spheroids for therapeutic treatments.
Collapse
Affiliation(s)
- Katherine H Griffin
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
- School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Shierly W Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
6
|
Petry F, Salzig D. The cultivation conditions affect the aggregation and functionality of β-cell lines alone and in coculture with mesenchymal stromal/stem cells. Eng Life Sci 2022; 22:769-783. [PMID: 36514533 PMCID: PMC9731603 DOI: 10.1002/elsc.202100168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/16/2022] Open
Abstract
The manufacturing of viable and functional β-cell spheroids is required for diabetes cell therapy and drug testing. Mesenchymal stromal/stem cells (MSCs) are known to improve β-cell viability and functionality. We therefore investigated the aggregation behavior of three different β-cell lines (rat insulinoma-1 cell line [INS-1], mouse insulinoma-6 cell line [MIN6], and a cell line formed by the electrofusion of primary human pancreatic islets and PANC-1 cells [1.1B4]), two MSC types, and mixtures of β-cells and MSCs under different conditions. We screened several static systems to produce uniform β-cell and MSC spheroids, finding cell-repellent plates the most suitable. The three different β-cell lines differed in their aggregation behavior, spheroid size, and growth in the same static environment. We found no major differences in spheroid formation between primary MSCs and an immortalized MSC line, although both differed with regard to the aggregation behavior of the β-cell lines. All spheroids showed a reduced viability due to mass transfer limitations under static conditions. We therefore investigated three dynamic systems (shaking multi-well plates, spinner flasks, and shaking flasks). In shaking flasks, there were no β-cell-line-dependent differences in aggregation behavior, resulting in uniform and highly viable spheroids. We found that the aggregation behavior of the β-cell lines changed in a static coculture with MSCs. The β-cell/MSC coculture conditions must be refined to avoid a rapid segregation into distinct populations under dynamic conditions.
Collapse
Affiliation(s)
- Florian Petry
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences MittelhessenGiessenGermany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences MittelhessenGiessenGermany
| |
Collapse
|
7
|
Nami Y, Kiani A, Elieh‐Ali‐Komi D, Jafari M, Haghshenas B. Impacts of alginate–basil seed mucilage–prebiotic microencapsulation on the survival rate of the potential probiotic
Leuconostoc mesenteroides
ABRIINW
.
N18
in yogurt. INT J DAIRY TECHNOL 2022. [DOI: 10.1111/1471-0307.12909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Yousef Nami
- Department of Food Biotechnology Branch for Northwest and West Region Agricultural Biotechnology Research Institute of Iran Agricultural Research, Education and Extension Organization (AREEO) Tabriz Iran
| | - Amir Kiani
- Regenerative Medicine Research Center (RMRC) Health Technology Institute Kermanshah University of Medical Sciences Kermanshah Iran
| | - Daniel Elieh‐Ali‐Komi
- Regenerative Medicine Research Center (RMRC) Health Technology Institute Kermanshah University of Medical Sciences Kermanshah Iran
| | - Mahdieh Jafari
- Department of Animal, Marine and Aquatic Biology and Biotechnology Faculty of Life Sciences and Biotechnology Shahid Beheshti University, Evin Tehran Iran
| | - Babak Haghshenas
- Regenerative Medicine Research Center (RMRC) Health Technology Institute Kermanshah University of Medical Sciences Kermanshah Iran
| |
Collapse
|
8
|
Patel SN, Mathews CE, Chandler R, Stabler CL. The Foundation for Engineering a Pancreatic Islet Niche. Front Endocrinol (Lausanne) 2022; 13:881525. [PMID: 35600597 PMCID: PMC9114707 DOI: 10.3389/fendo.2022.881525] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022] Open
Abstract
Progress in diabetes research is hindered, in part, by deficiencies in current experimental systems to accurately model human pathophysiology and/or predict clinical outcomes. Engineering human-centric platforms that more closely mimic in vivo physiology, however, requires thoughtful and informed design. Summarizing our contemporary understanding of the unique and critical features of the pancreatic islet can inform engineering design criteria. Furthermore, a broad understanding of conventional experimental practices and their current advantages and limitations ensures that new models address key gaps. Improving beyond traditional cell culture, emerging platforms are combining diabetes-relevant cells within three-dimensional niches containing dynamic matrices and controlled fluidic flow. While highly promising, islet-on-a-chip prototypes must evolve their utility, adaptability, and adoptability to ensure broad and reproducible use. Here we propose a roadmap for engineers to craft biorelevant and accessible diabetes models. Concurrently, we seek to inspire biologists to leverage such tools to ask complex and nuanced questions. The progenies of such diabetes models should ultimately enable investigators to translate ambitious research expeditions from benchtop to the clinic.
Collapse
Affiliation(s)
- Smit N. Patel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Rachel Chandler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
9
|
Kim MJ, Hwang YH, Hwang JW, Alam Z, Lee DY. Heme oxygenase-1 gene delivery for altering high mobility group box-1 protein in pancreatic islet. J Control Release 2022; 343:326-337. [PMID: 35085698 DOI: 10.1016/j.jconrel.2022.01.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
Abstract
Pancreatic islet transplantation is a promising strategy for the treatment of type I diabetes. High-mobility group box-1 (HMGB1), highly expressed in islet cells, is a potent immune stimulator in immune rejection. Heme oxygenase-1 (HO1) gene therapy can modulate the release of HMGB1 by altering intracellular molecules for successful cell transplantation. After delivery of the heme oxygenase-1 (HO1) gene to islet cells using an adeno-associated viral vector (AAV), it was evaluated the changes in cytoplasmic Ca2+ ions and calcineurin activity as well as histone acetyltransferase (HAT) and Poly(ADP) ribose polymerase-1 (PARP-1). Inhibition of HMGB1 release was evaluated through altering these intracellular molecules. Then, after transplantation of HO1-transduced islets, the therapeutic effect of them was evaluated through measuring blood glucose level to diabetic mice and through immunohistochemical analysis. The transduced HO1 gene significantly inhibited HMGB1 release in islets that was under the cell damage by hypoxia exposure. It was confirmed that this result was initially due to the decrease in cytoplasmic Ca2+ ion concentration and calcineurin activity. In addition, the delivered HO1 gene simultaneously reduced the activity of HAT and PARP-1, which are involved in the translocation of HMGB1 from the nucleus to the cytoplasm. As a result, when the HO1 gene-transduced islets were transplanted into diabetic mice, the treatment efficiency of diabetes was effectively improved by increasing the survival rate of the islets. Collectively, these results suggest that HO1 gene transfer can be used for successful islet transplantation by altering the activity of intracellular signal molecules and reducing HMGB1 release.
Collapse
Affiliation(s)
- Min Jun Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Yong Hwa Hwang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Jin Wook Hwang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Zahid Alam
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science & Technology (INST), Hanyang University, Seoul 04763, Republic of Korea; Elixir Pharmatech Inc., Seoul 04763, Republic of Korea.
| |
Collapse
|
10
|
Polymer nanotherapeutics to correct autoimmunity. J Control Release 2022; 343:152-174. [PMID: 34990701 DOI: 10.1016/j.jconrel.2021.12.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/11/2022]
Abstract
The immune system maintains homeostasis and protects the body from pathogens, mutated cells, and other harmful substances. When immune homeostasis is disrupted, excessive autoimmunity will lead to diseases. To inhibit the unexpected immune responses and reduce the impact of treatment on immunoprotective functions, polymer nanotherapeutics, such as nanomedicines, nanovaccines, and nanodecoys, were developed as part of an advanced strategy for precise immunomodulation. Nanomedicines transport cytotoxic drugs to target sites to reduce the occurrence of side effects and increase the stability and bioactivity of various immunomodulating agents, especially nucleic acids and cytokines. In addition, polymer nanomaterials carrying autoantigens used as nanovaccines can induce antigen-specific immune tolerance without interfering with protective immune responses. The precise immunomodulatory function of nanovaccines has broad prospects for the treatment of immune related-diseases. Besides, nanodecoys, which are designed to protect the body from various pathogenic substances by intravenous administration, are a simple and relatively noninvasive treatment. Herein, we have discussed and predicted the application of polymer nanotherapeutics in the correction of autoimmunity, including treating autoimmune diseases, controlling hypersensitivity, and avoiding transplant rejection.
Collapse
|
11
|
Liu H, Yang X, Cheng X, Zhao G, Zheng G, Li X, Dong R. Theoretical and Experimental Research on Multi-Layer Vessel-like Structure Printing Based on 3D Bio-Printing Technology. MICROMACHINES 2021; 12:mi12121517. [PMID: 34945366 PMCID: PMC8709219 DOI: 10.3390/mi12121517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Traditional autologous transplantation has become a severe issue due to insufficient donors. Artificial blood vessel is an effective method for the treatment of major vascular diseases, such as heart and peripheral blood vessel diseases. However, the traditional single-material printing technology has been unable to meet the users’ demand for product functional complexity, which is not only reflected in the field of industrial manufacturing, but also in the field of functional vessel-like structure regeneration. In order to achieve the printing and forming of multi-layer vessel-like structures, this paper carries out theoretical and experimental research on the printing and forming of a multi-layer vessel-like structure based on multi-material 3D bioprinting technology. Firstly, theoretical analysis has been explored to research the relationship among the different parameters in the process of vessel forming, and further confirm the synchronous relationship among the extrusion rate of material, the tangential speed of the rotating rod, and the movement speed of the platform. Secondly, sodium alginate and gelatin have been used as the experimental materials to manufacture the vessel-like structure, and the corrected parameter of the theoretical analysis is further verified. Finally, the cell-loaded materials have been printed and analyzed, and cell viability is more than 90%, which provides support for the research of multi-layer vessel-like structure printing.
Collapse
|
12
|
Aghlara-Fotovat S, Nash A, Kim B, Krencik R, Veiseh O. Targeting the extracellular matrix for immunomodulation: applications in drug delivery and cell therapies. Drug Deliv Transl Res 2021; 11:2394-2413. [PMID: 34176099 DOI: 10.1007/s13346-021-01018-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.
Collapse
Affiliation(s)
| | - Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Len’shina NA, Konev AN, Baten’kin AA, Bardina PS, Cherkasova EI, Kashina AV, Zagainova EV, Zagainov VE, Chesnokov SA. Alginate Functionalization for the Microencapsulation of Insulin Producing Cells. POLYMER SCIENCE SERIES B 2021. [DOI: 10.1134/s1560090421060129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Kaviani M, Keshtkar S, Sarvestani FS, Azarpira N, Yaghobi R, Aghdaei MH, Geramizadeh B, Esfandiari E, Shamsaeefar A, Nikeghbalian S, Al-Abdullah IH, Karimi MH, Motazedian N. The potential of the incorporated collagen microspheres in alginate hydrogel as an engineered three-dimensional microenvironment to attenuate apoptosis in human pancreatic islets. Acta Histochem 2021; 123:151775. [PMID: 34450327 DOI: 10.1016/j.acthis.2021.151775] [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: 04/21/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Tissue engineering is considered as a promising tool for remodeling the native cells microenvironment. In the present study, the effect of alginate hydrogel and collagen microspheres integrated with extracellular matrix components were evaluated in the decrement of apoptosis in human pancreatic islets. MATERIALS/METHODS For three-dimensional culture, the islets were encapsulated in collagen microspheres, containing laminin and collagen IV and embedded in alginate scaffold for one week. After that the islets were examined in terms of viability, apoptosis, genes and proteins expression including BAX, BCL2, active caspase-3, and insulin. Moreover, the islets function was evaluated through glucose-induced insulin and C-peptide secretion assay. In order to evaluate the structure of the scaffolds and the morphology of the pancreatic islets in three-dimensional microenvironments, we performed scanning electron microscopy. RESULTS Our findings showed that the designed hydrogel scaffolds significantly improved the islets viability using the reduction of activated caspase-3 and TUNEL positive cells. CONCLUSIONS The reconstruction of the destructed matrix with alginate hydrogels and collagen microspheres might be an effective step to promote the culture of the islets.
Collapse
Affiliation(s)
- Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Keshtkar
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ramin Yaghobi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Bita Geramizadeh
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elaheh Esfandiari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Shamsaeefar
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Nikeghbalian
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, USA
| | | | - Nasrin Motazedian
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Osteoclast-Mediated Cell Therapy as an Attempt to Treat Elastin Specific Vascular Calcification. Molecules 2021; 26:molecules26123643. [PMID: 34203711 PMCID: PMC8232296 DOI: 10.3390/molecules26123643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
Inflammation and stiffness in the arteries is referred to as vascular calcification. This process is a prevalent yet poorly understood consequence of cardiovascular disease and diabetes mellitus, comorbidities with few treatments clinically available. Because this is an active process similar to bone formation, it is hypothesized that osteoclasts (OCs), bone-resorbing cells in the body, could potentially work to reverse existing calcification by resorbing bone material. The receptor activator of nuclear kappa B-ligand (RANKL) is a molecule responsible for triggering a response in monocytes and macrophages that allows them to differentiate into functional OCs. In this study, OC and RANKL delivery were employed to determine whether calcification could be attenuated. OCs were either delivered via direct injection, collagen/alginate microbeads, or collagen gel application, while RANKL was delivered via injection, through either a porcine subdermal model or aortic injury model. While in vitro results yielded a decrease in calcification using OC therapy, in vivo delivery mechanisms did not provide control or regulation to keep cells localized long enough to induce calcification reduction. However, these results do provide context and direction for the future of OC therapy, revealing necessary steps for this treatment to effectively reduce calcification in vivo. The discrepancy between in vivo and in vitro success for OC therapy points to the need for a more stable and time-controlled delivery mechanism that will allow OCs not only to remain at the site of calcification, but also to be regulated so that they are healthy and functioning normally when introduced to diseased tissue.
Collapse
|
16
|
Hu T, Lo ACY. Collagen-Alginate Composite Hydrogel: Application in Tissue Engineering and Biomedical Sciences. Polymers (Basel) 2021; 13:1852. [PMID: 34199641 PMCID: PMC8199729 DOI: 10.3390/polym13111852] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Alginate (ALG), a polysaccharide derived from brown seaweed, has been extensively investigated as a biomaterial not only in tissue engineering but also for numerous biomedical sciences owing to its wide availability, good compatibility, weak cytotoxicity, low cost, and ease of gelation. Nevertheless, alginate lacks cell-binding sites, limiting long-term cell survival and viability in 3D culture. Collagen (Col), a major component protein found in the extracellular matrix (ECM), exhibits excellent biocompatibility and weak immunogenicity. Furthermore, collagen contains cell-binding motifs, which facilitate cell attachment, interaction, and spreading, consequently maintaining cell viability and promoting cell proliferation. Recently, there has been a growing body of investigations into collagen-based hydrogel trying to overcome the poor mechanical properties of collagen. In particular, collagen-alginate composite (CAC) hydrogel has attracted much attention due to its excellent biocompatibility, gelling under mild conditions, low cytotoxicity, controllable mechanic properties, wider availability as well as ease of incorporation of other biomaterials and bioactive agents. This review aims to provide an overview of the properties of alginate and collagen. Moreover, the application of CAC hydrogel in tissue engineering and biomedical sciences is also discussed.
Collapse
Affiliation(s)
| | - Amy C. Y. Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
17
|
Velasco-Mallorquí F, Rodríguez-Comas J, Ramón-Azcón J. Cellulose-based scaffolds enhance pseudoislets formation and functionality. Biofabrication 2021; 13. [PMID: 34075893 DOI: 10.1088/1758-5090/ac00c3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022]
Abstract
In vitroresearch for the study of type 2 diabetes (T2D) is frequently limited by the availability of a functional model for islets of Langerhans. To overcome the limitations of obtaining pancreatic islets from different sources, such as animal models or human donors, immortalized cell lines as the insulin-producing INS1Eβ-cells have appeared as a valid alternative to model insulin-related diseases. However, immortalized cell lines are mainly used in flat surfaces or monolayer distributions, not resembling the spheroid-like architecture of the pancreatic islets. To generate islet-like structures, the use of scaffolds appeared as a valid tool to promote cell aggregations. Traditionally-used hydrogel encapsulation methods do not accomplish all the requisites for pancreatic tissue engineering, as its poor nutrient and oxygen diffusion induces cell death. Here, we use cryogelation technology to develop a more resemblance scaffold with the mechanical and physical properties needed to engineer pancreatic tissue. This study shows that carboxymethyl cellulose (CMC) cryogels prompted cells to generateβ-cell clusters in comparison to gelatin-based scaffolds, that did not induce this cell organization. Moreover, the high porosity achieved with CMC cryogels allowed us to create specific range pseudoislets. Pseudoislets formed within CMC-scaffolds showed cell viability for up to 7 d and a better response to glucose over conventional monolayer cultures. Overall, our results demonstrate that CMC-scaffolds can be used to control the organization and function of insulin-producingβ-cells, representing a suitable technique to generateβ-cell clusters to study pancreatic islet function.
Collapse
Affiliation(s)
- Ferran Velasco-Mallorquí
- Biosensors for Bioengineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, Barcelona 08028, Spain
| | - Júlia Rodríguez-Comas
- Biosensors for Bioengineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, Barcelona 08028, Spain
| | - Javier Ramón-Azcón
- Biosensors for Bioengineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, Barcelona 08028, Spain.,ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| |
Collapse
|
18
|
Kahieshesfandiari M, Nami Y, Lornezhad G, Kiani A, Javanmard A, Jaymand M, Haghshenas B. Herbal hydrogel-based encapsulated Enterococcus faecium ABRIINW.N7 improves the resistance of red hybrid tilapia against Streptococcus iniae. J Appl Microbiol 2021; 131:2516-2527. [PMID: 33817937 DOI: 10.1111/jam.15098] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/06/2021] [Accepted: 03/29/2021] [Indexed: 01/19/2023]
Abstract
AIMS The streptococcal disease has been associated with serious mortality and significant global economic loss in the tilapia farming industry. The overall goal of this work was to test herbal hydrogels based on encapsulated Enterococcus faecium ABRIINW.N7 for potential probiotic anti-microbial activity against Streptococcus iniae in red hybrid tilapia. METHODS AND RESULTS Abnormal behaviour, clinical signs, postinjection survival and histopathology (kidney, liver, eye and brain) were measured. Cumulative mortality of CON+ , free cells, ALG and treatments (F1-F7) was 30, 24, 22, 19, 17, 17, 16, 14, 14 and 12 out of 30 fish and the survival rates for E. faecium ABRIINW.N7 microencapsulated in an alginate-BS blend with 0·5, 1, 1·5, 2, 2·5 and 3% fenugreek were 43, 43, 47, 53, 53 and 60%, respectively. After the incorporation of fenugreek with the alginate-BS blend, there was an 8-21% increase in probiotic cell viability. Furthermore, the survival rate for the alginate-BS blend with 2·5 and 3% fenugreek (F6 and F7) was significantly (P ≤ 0·05) higher than other blends. The highest encapsulation efficiency, viability in gastrointestinal conditions and during storage time and excellent antipathogenicity against S. iniae were observed in alginate-BS +3% fenugreek formulation (F7). CONCLUSIONS It is recommended that probiotic strains like E. faecium ABRIINW.N7 in combination with local herbal gums, such as BS and fenugreek plus alginate, can be used as a suitable scaffold and an ideal matrix for the encapsulation of probiotics. SIGNIFICANCE AND IMPACT OF THE STUDY This study proposes models connecting process parameters, matrix structure and functionality.
Collapse
Affiliation(s)
- M Kahieshesfandiari
- Department of Aquaculture, Faculty of Agriculture, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - Y Nami
- Department of Food Biotechnology, Branch for Northwest and West Region, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO), Tabriz, Iran
| | - G Lornezhad
- School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - A Kiani
- Regenerative Medicine Research Center (RMRC), Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - A Javanmard
- Animal Genetics and Breeding Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - M Jaymand
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - B Haghshenas
- Regenerative Medicine Research Center (RMRC), Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
19
|
Treatment of diabetic mice by microfluidic system-assisted transplantation of stem cells-derived insulin-producing cells transduced with miRNA. Life Sci 2021; 274:119338. [PMID: 33716064 DOI: 10.1016/j.lfs.2021.119338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 01/22/2023]
Abstract
AIMS Cell-based therapy is a promising approach for the treatment of type-1 diabetes mellitus. Identifying stem cells with differentiation potential to Insulin-producing cells (IPCs) and their application is an emerging issue. Different strategies have been used to support cell survival and their specific functions to control hyperglycemia conditions. Novel technologies using appropriate materials/fibers can improve cell transplantation. MAIN METHODS In the present study, IPCs were differentiated from adipose-derived stem cells transduced with miR-375 and anti-miR-7. The cells' survival rate was also improved using a microfluidic system before their in vivo transplantation. KEY FINDINGS After adopting a stable, functional condition of the IPCs, the cells were used for in vivo grafting to diabetic mice, which resulted in a substantial drop in blood glucose during four weeks of grafting compared to the control group (p < 0.0001). The pattern of blood glucose levels in the mice receiving fiber entrapped IPCs, was similar to that of non-diabetic mice. Blood insulin was elevated in diabetic mice which received a transplant of fiber-entrapped-IPCs carrying miR-375 and anti-miR-7 after five weeks of transplantation compared to the diabetic mice (p < 0.014). SIGNIFICANCE For the first time, this study showed that the two-component microfluidic system is useful for supporting the Collagen-Alginate fiber-entrapped IPCs and the miRNA-based cell therapy. Overall, our data show that the IPC encapsulation using a microfluidic system can support the cells in terms of morphology and biological function and their efficiency for controlling the hyperglycemia condition in diabetic mice.
Collapse
|
20
|
Enck K, Tamburrini R, Deborah C, Gazia C, Jost A, Khalil F, Alwan A, Orlando G, Opara EC. Effect of alginate matrix engineered to mimic the pancreatic microenvironment on encapsulated islet function. Biotechnol Bioeng 2020; 118:1177-1185. [PMID: 33270214 DOI: 10.1002/bit.27641] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/15/2020] [Accepted: 11/21/2020] [Indexed: 12/13/2022]
Abstract
Islet transplantation is emerging as a therapeutic option for type 1 diabetes, albeit, only a small number of patients meeting very stringent criteria are eligible for the treatment because of the side effects of the necessary immunosuppressive therapy and the relatively short time frame of normoglycemia that most patients achieve. The challenge of the immune-suppressive regimen can be overcome through microencapsulation of the islets in a perm-selective coating of alginate microbeads with poly-l-lysine or poly- l-ornithine. In addition to other issues including the nutrient supply challenge of encapsulated islets a critical requirement for these cells has emerged as the need to engineer the microenvironment of the encapsulation matrix to mimic that of the native pancreatic scaffold that houses islet cells. That microenvironment includes biological and mechanical cues that support the viability and function of the cells. In this study, the alginate hydrogel was modified to mimic the pancreatic microenvironment by incorporation of extracellular matrix (ECM). Mechanical and biological changes in the encapsulating alginate matrix were made through stiffness modulation and incorporation of decellularized ECM, respectively. Islets were then encapsulated in this new biomimetic hydrogel and their insulin production was measured after 7 days in vitro. We found that manipulation of the alginate hydrogel matrix to simulate both physical and biological cues for the encapsulated islets enhances the mechanical strength of the encapsulated islet constructs as well as their function. Our data suggest that these modifications have the potential to improve the success rate of encapsulated islet transplantation.
Collapse
Affiliation(s)
- Kevin Enck
- Wake Forest School of Medicine, Virginia Tech School of Biomedical Engineering & Sciences (SBES), Wake Forest University, Winston-Salem, North Carolina.,Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| | - Riccardo Tamburrini
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Chaimov Deborah
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Carlo Gazia
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Alec Jost
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Fatma Khalil
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| | - Abdelrahman Alwan
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| | - Giuseppe Orlando
- Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina.,Department of Surgery, Wake Forest University, Winston-Salem, North Carolina
| | - Emmanuel C Opara
- Wake Forest School of Medicine, Virginia Tech School of Biomedical Engineering & Sciences (SBES), Wake Forest University, Winston-Salem, North Carolina.,Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine (WFIRM), Winston-Salem, North Carolina
| |
Collapse
|
21
|
Wang JK, Cheam NMJ, Irvine SA, Tan NS, Venkatraman S, Tay CY. Interpenetrating Network of Alginate–Human Adipose Extracellular Matrix Hydrogel for Islet Cells Encapsulation. Macromol Rapid Commun 2020; 41:e2000275. [DOI: 10.1002/marc.202000275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/04/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Jun Kit Wang
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
| | - Nicole Mein Ji Cheam
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
| | - Scott Alexander Irvine
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
| | - Nguan Soon Tan
- School of Biological Sciences Nanyang Technological University Singapore 60 Nanyang Drive Singapore 637551 Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore 11 Mandalay Road Singapore 308232 Singapore
| | - Subbu Venkatraman
- Department of Materials Science and Engineering National University of Singapore Blk EA, 9 Engineering Drive 1 Singapore 117575 Singapore
| | - Chor Yong Tay
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
- School of Biological Sciences Nanyang Technological University Singapore 60 Nanyang Drive Singapore 637551 Singapore
- Environmental Chemistry and Materials Centre Nanyang Environment and Water Research Institute 1 CleanTech Loop, CleanTech One Singapore 637141 Singapore
| |
Collapse
|
22
|
Watanabe T, Okitsu T, Ozawa F, Nagata S, Matsunari H, Nagashima H, Nagaya M, Teramae H, Takeuchi S. Millimeter-thick xenoislet-laden fibers as retrievable transplants mitigate foreign body reactions for long-term glycemic control in diabetic mice. Biomaterials 2020; 255:120162. [PMID: 32562943 DOI: 10.1016/j.biomaterials.2020.120162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 05/19/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Transplantation technologies of pancreatic islets as well as stem cell-derived pancreatic beta cells encapsulated in hydrogel for the induction of immunoprotection could advance to treat type 1 diabetes mellitus, if the hydrogel transplants acquire retrievability through mitigating foreign body reactions after transplantation. Here, we demonstrate that the diameter of the fiber-shaped hydrogel transplants determines both in vivo cellular deposition onto themselves and their retrievability. Specifically, we found that the in vivo cellular deposition is significantly mitigated when the diameter is 1.0 mm and larger, and that 1.0 mm-thick xenoislet-laden fiber-shaped hydrogel transplants can be retrieved after being placed in the intraperitoneal cavities of immunocompetent diabetic mice for more than 100 days, during which period the hydrogel transplants can normalize the blood glucose concentrations of the mice. These findings could provide an innovative concept of a transplant that would promote the clinical application of stem cell-derived functional cells through improving their in vivo efficacy and safety.
Collapse
Affiliation(s)
- Takaichi Watanabe
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Teru Okitsu
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Fumisato Ozawa
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Shogo Nagata
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan; Laboratory of Developmental Engineering, Meiji University, Kawasaki, 214-8571, Japan
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan
| | - Hiroki Teramae
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Shoji Takeuchi
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
23
|
Nami Y, Lornezhad G, Kiani A, Abdullah N, Haghshenas B. Alginate-Persian Gum-Prebiotics microencapsulation impacts on the survival rate of Lactococcus lactis ABRIINW-N19 in orange juice. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109190] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Bonani W, Cagol N, Maniglio D. Alginate Hydrogels: A Tool for 3D Cell Encapsulation, Tissue Engineering, and Biofabrication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1250:49-61. [PMID: 32601937 DOI: 10.1007/978-981-15-3262-7_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A wide variety of hydrogels have been proposed for tissue engineering applications, cell encapsulation, and bioinks for bioprinting applications. Cell-laden hydrogel constructs rely on natural hydrogels such as alginate, agarose, chitosan, collagen, gelatin, fibroin, and hyaluronic acid (HA), as well as on synthetic hydrogels such as poloxamers (Pluronics®) and polyethylene glycol (PEG). Alginate has become more and more important in the last years, thanks to the possibility to prepare alginate hydrogels suitable for cell encapsulation mainly because of the mild and reversible cross-linking conditions. In this paper alginate will be described in detail with respect to its chemistry, cross-linking behavior, biocompatibility, manufacturing capacity, and possible modifications.
Collapse
Affiliation(s)
- Walter Bonani
- Directorate for Nuclear Safety and Security, European Commission, Joint Research Centre, Karlsruhe, Germany.,Department of Industrial Engineering and BIOtech Research Center, University of Trento, Trento, Italy
| | - Nicola Cagol
- Department of Industrial Engineering and BIOtech Research Center, University of Trento, Trento, Italy
| | - Devid Maniglio
- Department of Industrial Engineering and BIOtech Research Center, University of Trento, Trento, Italy.
| |
Collapse
|
25
|
Cheng Y, Zhang Y, Yu Y, Zhao G, Zhao Y, He X. Cold-Responsive Nanocapsules Enable the Sole-Cryoprotectant-Trehalose Cryopreservation of β Cell-Laden Hydrogels for Diabetes Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904290. [PMID: 31833664 DOI: 10.1002/smll.201904290] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/17/2019] [Indexed: 05/27/2023]
|
26
|
Design and manufacture of 3D cell culture plate for mass production of cell-spheroids. Sci Rep 2019; 9:13976. [PMID: 31562370 PMCID: PMC6765015 DOI: 10.1038/s41598-019-50186-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
A 3D cell culture is preferred to 2D cell culture since it allows cells to grow in all directions in vitro, similar to how they would in vivo. 3D cell culture plates currently used in tissue engineering research have limited access to control the geometry. Furthermore, 3D cell culture plate manufacturing methods are relatively complex, time-consuming, labor-intensive, and expensive. Therefore, a design and manufacturing method, which has relatively low cost, high throughput, and high size flexibility, is proposed. Cell culture plate was fabricated by computer aided design and manufacturing software using polydimethylsiloxane as a plate constituent. With the successfully-developed 3D cell culture plate, the morphology and viability of the cultured mesenchymal stem cells were tested.The mesenchymal stem cells seeded on the newly-fabricated 3D cell culture plate aggregated to form 3D spheroids within 24 h of incubation and well-maintained their viability. Thus, due to the capacity of mass production of the cell spheroids with a desired cell viability, the newly-fabricated plate has a great promise to prepare 3D cell spheroids for experimental as well as clinical applications.
Collapse
|
27
|
3D-Models of Insulin-Producing β-Cells: from Primary Islet Cells to Stem Cell-Derived Islets. Stem Cell Rev Rep 2018; 14:177-188. [PMID: 29181780 DOI: 10.1007/s12015-017-9783-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a need for physiologically relevant assay platforms to provide functionally relevant models of diabetes, to accelerate the discovery of new treatment options and boost developments in drug discovery. In this review, we compare several 3D-strategies that have been used to increase the functional relevance of ex vivo human primary pancreatic islets and developments into the generation of stem cell derived pancreatic beta-cells (β-cells). Special attention will be given to recent approaches combining the use of extracellular matrix (ECM) scaffolds with pancreatic molecular memory, which can be used to improve yield and functionality of in vitro stem cell-derived pancreatic models. The ultimate goal is to develop scalable cell-based platforms for diabetes research and drug screening. This article will critically assess key aspects related to in vitro pancreatic 3D-ECM models and highlight the most promising approaches for future research.
Collapse
|
28
|
Stephens CH, Orr KS, Acton AJ, Tersey SA, Mirmira RG, Considine RV, Voytik-Harbin SL. In situ type I oligomeric collagen macroencapsulation promotes islet longevity and function in vitro and in vivo. Am J Physiol Endocrinol Metab 2018; 315:E650-E661. [PMID: 29894201 PMCID: PMC6230705 DOI: 10.1152/ajpendo.00073.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Widespread use of pancreatic islet transplantation for treatment of type 1 diabetes (T1D) is currently limited by requirements for long-term immunosuppression, limited donor supply, and poor long-term engraftment and function. Upon isolation from their native microenvironment, islets undergo rapid apoptosis, which is further exacerbated by poor oxygen and nutrient supply following infusion into the portal vein. Identifying alternative strategies to restore critical microenvironmental cues, while maximizing islet health and function, is needed to advance this cellular therapy. We hypothesized that biophysical properties provided through type I oligomeric collagen macroencapsulation are important considerations when designing strategies to improve islet survival, phenotype, and function. Mouse islets were encapsulated at various Oligomer concentrations (0.5 -3.0 mg/ml) or suspended in media and cultured for 14 days, after which viability, protein expression, and function were assessed. Oligomer-encapsulated islets showed a density-dependent improvement in in vitro viability, cytoarchitecture, and insulin secretion, with 3 mg/ml yielding values comparable to freshly isolated islets. For transplantation into streptozotocin-induced diabetic mice, 500 islets were mixed in Oligomer and injected subcutaneously, where rapid in situ macroencapsulation occurred, or injected with saline. Mice treated with Oligomer-encapsulated islets exhibited rapid (within 24 h) diabetes reversal and maintenance of normoglycemia for 14 (immunocompromised), 90 (syngeneic), and 40 days (allogeneic). Histological analysis showed Oligomer-islet engraftment with maintenance of islet cytoarchitecture, revascularization, and no foreign body response. Oligomer-islet macroencapsulation may provide a useful strategy for prolonging the health and function of cultured islets and has potential as a subcutaneous injectable islet transplantation strategy for treatment of T1D.
Collapse
Affiliation(s)
| | - Kara S Orr
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Pediatrics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Anthony J Acton
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sarah A Tersey
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Pediatrics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Raghavendra G Mirmira
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Pediatrics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Robert V Considine
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University , West Lafayette, Indiana
- Department of Basic Medical Sciences, Purdue University , West Lafayette, Indiana
| |
Collapse
|
29
|
Choe G, Park J, Park H, Lee JY. Hydrogel Biomaterials for Stem Cell Microencapsulation. Polymers (Basel) 2018; 10:E997. [PMID: 30960922 PMCID: PMC6403586 DOI: 10.3390/polym10090997] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
Stem cell transplantation has been recognized as a promising strategy to induce the regeneration of injured and diseased tissues and sustain therapeutic molecules for prolonged periods in vivo. However, stem cell-based therapy is often ineffective due to low survival, poor engraftment, and a lack of site-specificity. Hydrogels can offer several advantages as cell delivery vehicles, including cell stabilization and the provision of tissue-like environments with specific cellular signals; however, the administration of bulk hydrogels is still not appropriate to obtain safe and effective outcomes. Hence, stem cell encapsulation in uniform micro-sized hydrogels and their transplantation in vivo have recently garnered great attention for minimally invasive administration and the enhancement of therapeutic activities of the transplanted stem cells. Several important methods for stem cell microencapsulation are described in this review. In addition, various natural and synthetic polymers, which have been employed for the microencapsulation of stem cells, are reviewed in this article.
Collapse
Affiliation(s)
- Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
| | - Junha Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
| |
Collapse
|
30
|
Kim H, Cho CH, Park JK. High-throughput culture and embedment of spheroid array using droplet contact-based spheroid transfer. BIOMICROFLUIDICS 2018; 12:044109. [PMID: 30867862 PMCID: PMC6404923 DOI: 10.1063/1.5039965] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/01/2018] [Indexed: 05/17/2023]
Abstract
Spheroids are one of the most representative models of 3D cell culture, which can be easily formed using conventional hanging drop method. However, medium change and spheroid transferring process are the bottlenecks that reduce the throughput of the entire process in the hanging drop culture. In addition, the embedment of spheroid into hydrogel still depends on the individual pipetting process. To overcome these issues, we present poly(dimethylsiloxane) (PDMS)-based simple devices which can exploit droplet contact-based spheroid transfer using a drop array chip (DAC) having an array of well structures and peripheral rims. When the upper spheroid-containing drops were in contact with the lower liquid drops, the air-liquid interface disappeared at the merged surface and the spheroids settled down due to gravitational force. This method was applied to repetitive medium change and live/dead staining of spheroids cultured with the hanging drop method. To simultaneously embed the spheroids into the corresponding collagen hydrogel drops, a PDMS-based pillar array chip (PAC) was contacted in advance with the spheroid-containing DAC. The contacted PAC then contained the spheroids trapped in small drops of liquid reduced in volume to around 0.5 μl. Consequently, the spheroids were embedded into the collagen drops at once by contacting the spheroid-containing PAC with the collagen-loaded DAC. The embedded spheroids using the DAC-PAC contacting method showed a reliable invasion behavior compared to the embedded spheroids using conventional manual pipetting.
Collapse
Affiliation(s)
- Hwisoo Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Chang Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Je-Kyun Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
31
|
Kim Y, Baipaywad P, Jeong Y, Park H. Incorporation of gelatin microparticles on the formation of adipose-derived stem cell spheroids. Int J Biol Macromol 2018; 110:472-478. [DOI: 10.1016/j.ijbiomac.2018.01.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/22/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
|
32
|
Petry F, Weidner T, Czermak P, Salzig D. Three-Dimensional Bioreactor Technologies for the Cocultivation of Human Mesenchymal Stem/Stromal Cells and Beta Cells. Stem Cells Int 2018; 2018:2547098. [PMID: 29731775 PMCID: PMC5872596 DOI: 10.1155/2018/2547098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a prominent health problem caused by the failure of pancreatic beta cells. One therapeutic approach is the transplantation of functional beta cells, but it is difficult to generate sufficient beta cells in vitro and to ensure these cells remain viable at the transplantation site. Beta cells suffer from hypoxia, undergo apoptosis, or are attacked by the host immune system. Human mesenchymal stem/stromal cells (hMSCs) can improve the functionality and survival of beta cells in vivo and in vitro due to direct cell contact or the secretion of trophic factors. Current cocultivation concepts with beta cells are simple and cannot exploit the favorable properties of hMSCs. Beta cells need a three-dimensional (3D) environment to function correctly, and the cocultivation setup is therefore more complex. This review discusses 3D cultivation forms (aggregates, capsules, and carriers) for hMSCs and beta cells and strategies for large-scale cultivation. We have determined process parameters that must be balanced and considered for the cocultivation of hMSCs and beta cells, and we present several bioreactor setups that are suitable for such an innovative cocultivation approach. Bioprocess engineering of the cocultivation processes is necessary to achieve successful beta cell therapy.
Collapse
Affiliation(s)
- Florian Petry
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| | - Tobias Weidner
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
- Department of Chemical Engineering, Kansas State University, Manhattan, KS, USA
- Project Group Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Winchesterstr. 3, 35394 Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| |
Collapse
|
33
|
Lee G, Jun Y, Jang H, Yoon J, Lee J, Hong M, Chung S, Kim DH, Lee S. Enhanced oxygen permeability in membrane-bottomed concave microwells for the formation of pancreatic islet spheroids. Acta Biomater 2018; 65:185-196. [PMID: 29101017 DOI: 10.1016/j.actbio.2017.10.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/22/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022]
Abstract
Oxygen availability is a critical factor in regulating cell viability that ultimately contributes to the normal morphogenesis and functionality of human tissues. Among various cell culture platforms, construction of 3D multicellular spheroids based on microwell arrays has been extensively applied to reconstitute in vitro human tissue models due to its precise control of tissue culture conditions as well as simple fabrication processes. However, an adequate supply of oxygen into the spheroidal cellular aggregation still remains one of the main challenges to producing healthy in vitro spheroidal tissue models. Here, we present a novel design for controlling the oxygen distribution in concave microwell arrays. We show that oxygen permeability into the microwell is tightly regulated by varying the poly-dimethylsiloxane (PDMS) bottom thickness of the concave microwells. Moreover, we validate the enhanced performance of the engineered microwell arrays by culturing non-proliferated primary rat pancreatic islet spheroids on varying bottom thickness from 10 μm to 1050 μm. Morphological and functional analyses performed on the pancreatic islet spheroids grown for 14 days prove the long-term stability, enhanced viability, and increased hormone secretion under the sufficient oxygen delivery conditions. We expect our results could provide knowledge on oxygen distribution in 3-dimensional spheroidal cell structures and critical design concept for tissue engineering applications. STATEMENT OF SIGNIFICANCE In this study, we present a noble design to control the oxygen distribution in concave microwell arrays for the formation of highly functional pancreatic islet spheroids by engineering the bottom of the microwells. Our new platform significantly enhanced oxygen permeability that turned out to improve cell viability and spheroidal functionality compared to the conventional thick-bottomed 3-D culture system. Therefore, we believe that this could be a promising medical biotechnology platform to further develop high-throughput tissue screening system as well as in vivo-mimicking customised 3-D tissue culture systems.
Collapse
|
34
|
Thomas D, O'Brien T, Pandit A. Toward Customized Extracellular Niche Engineering: Progress in Cell-Entrapment Technologies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1703948. [PMID: 29194781 DOI: 10.1002/adma.201703948] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/12/2017] [Indexed: 06/07/2023]
Abstract
The primary aim in tissue engineering is to repair, replace, and regenerate dysfunctional tissues to restore homeostasis. Cell delivery for repair and regeneration is gaining impetus with our understanding of constructing tissue-like environments. However, the perpetual challenge is to identify innovative materials or re-engineer natural materials to model cell-specific tissue-like 3D modules, which can seamlessly integrate and restore functions of the target organ. To devise an optimal functional microenvironment, it is essential to define how simple is complex enough to trigger tissue regeneration or restore cellular function. Here, the purposeful transition of cell immobilization from a cytoprotection point of view to that of a cell-instructive approach is examined, with advances in the understanding of cell-material interactions in a 3D context, and with a view to further application of the knowledge for the development of newer and complex hierarchical tissue assemblies for better examination of cell behavior and offering customized cell-based therapies for tissue engineering.
Collapse
Affiliation(s)
- Dilip Thomas
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
35
|
Huang Y, Wang Y, Chen L, Zhang L. Facile construction of mechanically tough collagen fibers reinforced by chitin nanofibers as cell alignment templates. J Mater Chem B 2018; 6:918-929. [DOI: 10.1039/c7tb02945d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Reconstituted collagen fibers with excellent mechanical performance were successfully fabricated with sodium alginate as coagulate and chitin nanofibers as reinforcing filler and applied as a fibroblast alignment templated scaffold.
Collapse
Affiliation(s)
- Yao Huang
- Department of Agricultural
- Food and Nutritional Science
- University of Alberta
- Edmonton
- Canada
| | - Yixiang Wang
- Department of Agricultural
- Food and Nutritional Science
- University of Alberta
- Edmonton
- Canada
| | - Lingyun Chen
- Department of Agricultural
- Food and Nutritional Science
- University of Alberta
- Edmonton
- Canada
| | - Lina Zhang
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
- China
| |
Collapse
|
36
|
Long G, Zhang G, Zhang F, Li M, Ye D, Yang D, Yang Y. Cotransplantation of Mesenchymal Stem Cells and Immature Dendritic Cells Potentiates the Blood Glucose Control of Islet Allografts. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4107943. [PMID: 29410963 PMCID: PMC5749219 DOI: 10.1155/2017/4107943] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/23/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transplantation of islets is a promising alternative to treat type 1 diabetes (T1D), but graft rejection is the major obstacle to its application in clinical practice. We evaluated the effects of mesenchymal stem cells (MSCs) and immature dendritic cells (imDCs) on islet transplantation in diabetic model. METHODS The streptozotocin T1D model was established in BABL/c mice. Rat islets were isolated and identified with dithizone (DTZ) staining. MSCs and imDCs were isolated from bone marrow of syngenic mice. Islets, alone or along with MSCs and/or imDCs, were transplanted to the left kidney capsule of diabetic mice. The blood glucose levels and glycosylated hemoglobin levels after transplantation were monitored. RESULTS Cotransplantation significantly decreased blood glucose and glycosylated hemoglobin levels in the diabetes mice. Transplantation of 200 islets + 2 × 105 MSCs + 2 × 105 imDCs could not only restore normal blood glucose levels, but also significantly prolong graft survival for 12.6 ± 3.48 days. CONCLUSIONS Cotransplantation of allogenic islets with imDCs and/or MSCs can significantly promote graft survival, reverse hyperglycemia, and effectively control the glycosylated hemoglobin levels.
Collapse
Affiliation(s)
- Guanghui Long
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guangtao Zhang
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fangting Zhang
- Center Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Minghua Li
- Center Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dongshuo Ye
- Shenzhen BioScien Pharmaceuticals Co. LTD, Shenzhen, China
| | - Dengke Yang
- Shenzhen BioScien Pharmaceuticals Co. LTD, Shenzhen, China
| | - Yinke Yang
- Shenzhen BioScien Pharmaceuticals Co. LTD, Shenzhen, China
| |
Collapse
|
37
|
Ribeiro D, Andersson EM, Heath N, Persson-Kry A, Collins R, Hicks R, Dekker N, Forslöw A. Human pancreatic islet-derived extracellular vesicles modulate insulin expression in 3D-differentiating iPSC clusters. PLoS One 2017; 12:e0187665. [PMID: 29117231 PMCID: PMC5678888 DOI: 10.1371/journal.pone.0187665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/24/2017] [Indexed: 01/02/2023] Open
Abstract
It has been suggested that extracellular vesicles (EVs) can mediate crosstalk between hormones and metabolites within pancreatic tissue. However, the possible effect of pancreatic EVs on stem cell differentiation into pancreatic lineages remains unknown. Herein, human islet-derived EVs (h-Islet-EVs) were isolated, characterized and subsequently added to human induced pluripotent stem cell (iPSC) clusters during pancreatic differentiation. The h-islet-EVs had a mean size of 117±7 nm and showed positive expression of CD63 and CD81 EV markers as measured by ELISA. The presence of key pancreatic transcription factor mRNA, such as NGN3, MAFA and PDX1, and pancreatic hormone proteins such as C-peptide and glucagon, were confirmed in h-Islet-EVs. iPSC clusters were differentiated in suspension and at the end stages of the differentiation protocol, the mRNA expression of the main pancreatic transcription factors and pancreatic hormones was increased. H-Islet-EVs were supplemented to the iPSC clusters in the later stages of differentiation. It was observed that h-Islet-EVs were able to up-regulate the intracellular levels of C-peptide in iPSC clusters in a concentration-dependent manner. The effect of h-Islet-EVs on the differentiation of iPSC clusters cultured in 3D-collagen hydrogels was also assessed. Although increased mRNA expression for pancreatic markers was observed when culturing the iPSC clusters in 3D-collagen hydrogels, delivery of EVs did not affect the insulin or C-peptide intracellular content. Our results provide new information on the role of h-Islet-EVs in the regulation of insulin expression in differentiating iPSC clusters, and are highly relevant for pancreatic tissue engineering applications.
Collapse
Affiliation(s)
- Diana Ribeiro
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden.,Department of Biology and Bioengineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva-Marie Andersson
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Nikki Heath
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Anette Persson-Kry
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Richard Collins
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Ryan Hicks
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Niek Dekker
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anna Forslöw
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
38
|
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Around 90% of deaths are caused by metastasis and just 10% by primary tumor. The advancement of treatment approaches is not at the same rhythm of the disease; making cancer a focal target of biomedical research. To enhance the understanding and prompts the therapeutic delivery; concepts of tissue engineering are applied in the development of in vitro models that can bridge between 2D cell culture and animal models, mimicking tissue microenvironment. Tumor spheroid represents highly suitable 3D organoid-like framework elucidating the intra and inter cellular signaling of cancer, like that formed in physiological niche. However, spheroids are of limited value in studying critical biological phenomenon such as tumor-stroma interactions involving extra cellular matrix or immune system. Therefore, a compelling need of tailoring spheroid technologies with physiologically relevant biomaterials or in silico models, is ever emerging. The diagnostic and prognostic role of spheroids rearrangements within biomaterials or microfluidic channel is indicative of patient management; particularly for the decision of targeted therapy. Fragmented information on available in vitro spheroid models and lack of critical analysis on transformation aspects of these strategies; pushes the urge to comprehensively overview the recent technological advancements (e.g. bioprinting, micro-fluidic technologies or use of biomaterials to attain the third dimension) in the shed of translationable cancer research. In present article, relationships between current models and their possible exploitation in clinical success is explored with the highlight of existing challenges in defining therapeutic targets and screening of drug efficacy.
Collapse
|
39
|
Eter WA, Van der Kroon I, Andralojc K, Buitinga M, Willekens SMA, Frielink C, Bos D, Joosten L, Boerman OC, Brom M, Gotthardt M. Non-invasive in vivo determination of viable islet graft volume by 111In-exendin-3. Sci Rep 2017; 7:7232. [PMID: 28775287 PMCID: PMC5543140 DOI: 10.1038/s41598-017-07815-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/30/2017] [Indexed: 01/20/2023] Open
Abstract
Pancreatic islet transplantation is a promising therapy for patients with type 1 diabetes. However, the duration of long-term graft survival is limited due to inflammatory as well as non-inflammatory processes and routine clinical tests are not suitable to monitor islet survival. 111In-exendin-SPECT (single photon emission computed tomography) is a promising method to non-invasively image islets after transplantation and has the potential to help improve the clinical outcome. Whether 111In-exendin-SPECT allows detecting small differences in beta-cell mass (BCM) and measuring the actual volume of islets that were successfully engrafted has yet to be demonstrated. Here, we evaluated the performance of 111In-exendin-SPECT using an intramuscular islet transplantation model in C3H mice. In vivo imaging of animals transplanted with 50, 100, 200, 400 and 800 islets revealed an excellent linear correlation between SPECT quantification of 111In-exendin uptake and insulin-positive area of islet transplants, demonstrating that 111In-exendin-SPECT specifically and accurately measures BCM. The high sensitivity of the method allowed measuring small differences in graft volumes, including grafts that contained less than 50 islets. The presented method is reliable, convenient and holds great potential for non-invasive monitoring of BCM after islet transplantation in humans.
Collapse
Affiliation(s)
- Wael A Eter
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Inge Van der Kroon
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Karolina Andralojc
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mijke Buitinga
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Stefanie M A Willekens
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Cathelijne Frielink
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Desiree Bos
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Maarten Brom
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Yajima Y, Yamada M, Utoh R, Seki M. Collagen Microparticle-Mediated 3D Cell Organization: A Facile Route to Bottom-up Engineering of Thick and Porous Tissues. ACS Biomater Sci Eng 2017; 3:2144-2154. [DOI: 10.1021/acsbiomaterials.7b00131] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Yuya Yajima
- Department of Applied Chemistry
and Biotechnology, Graduate School of Engineering, Chiba University, 1-33
Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry
and Biotechnology, Graduate School of Engineering, Chiba University, 1-33
Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Rie Utoh
- Department of Applied Chemistry
and Biotechnology, Graduate School of Engineering, Chiba University, 1-33
Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Minoru Seki
- Department of Applied Chemistry
and Biotechnology, Graduate School of Engineering, Chiba University, 1-33
Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
41
|
Pancreatic Islet Transplantation Technologies: State of the Art of Micro- and Macro-Encapsulation. CURRENT TRANSPLANTATION REPORTS 2017. [DOI: 10.1007/s40472-017-0154-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
42
|
Park D, Park J, Jang H, Cheng J, Hyun Kim S, Lee SH. Simultaneous microfluidic spinning of multiple strands of submicron fiber for the production of free-standing porous membranes for biological application. Biofabrication 2017; 9:025026. [PMID: 28504242 DOI: 10.1088/1758-5090/aa7307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Microfibers produced using electrospinning and microfluidics-based technologies have been developed as a powerful tool in tissue engineering applications such as drug delivery and scaffolds. The applications of these fibers, however, have been limited because of the hazardous solvents used to make them, difficulties in controlling the pore sizes of their membrane forms, and downscaling the size of the fiber. Nevertheless, extending the use of these fibers, for example in the production of a free-standing porous membrane appropriate for cell-based research, is highly needed for tissue engineering, organ-on-a-chip, and drug delivery research and applications. Here, we fabricated a free-standing porous membrane by using a novel method that involved simultaneously spinning multiple strands of submicron-thick 'noodle-like' fibers. In addition to the novelty of the single noodle fiber in overcoming the size-reducing limitations of conventional microfluidic spinning methods, these fibers can hence form the units of 'noodle membranes' whose pores have sizes that the convention electrospinning method cannot achieve. We confirmed the potential of the noodle membrane to serve as a free-standing porous membrane in two simple experiments. Also, we found that noodle membranes have an advantage in loading different amounts of different materials in itself that it was also shown to be of use as a new type of scaffold for complex tissue regeneration. Therefore, the proposed noodle membrane can be an effective tool in tissue engineering applications and biological studies.
Collapse
Affiliation(s)
- DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | | | | | | | | | | |
Collapse
|
43
|
Bae CY, Son J, Kim H, Park JK. Demonstration of Interposed Modular Hydrogel Sheet for Multicellular Analysis in a Microfluidic Assembly Platform. Sci Rep 2017; 7:1289. [PMID: 28465532 PMCID: PMC5430983 DOI: 10.1038/s41598-017-01363-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/29/2017] [Indexed: 12/29/2022] Open
Abstract
Hydrogel sheets have emerged as a promising biomaterial scaffold for the encapsulation and transfer of multicellular structures. Although the improvement of the chemical interactions and the design of micro-scaled geometry have contributed to the development of multipurpose hydrogel scaffolds, the application of hydrogel sheets to assess multicellular structures is still challenging. To expand the technical applicability of hydrogel sheets, we here demonstrate that a single layer of the hydrogel sheet can be integrated as an interposed module in a microfluidic device for multicellular analysis. As a cell culture unit, encapsulated pancreatic insulinoma (MIN6) cells in the hydrogel sheet were labeled and examined via multiple microchannels. After obtaining simultaneously multi-labeled cells in the hydrogel sheet that had been incorporated into the microfluidic device, each modular hydrogel sheet was also recoverable and re-cultured without any distortion. The modular hydrogel sheet can be simply manipulated and conserved as a multicellular module in a three-dimensional (3D) in vitro culture platform. Using the modular concept of hydrogel sheets capable of cell culture and/or assay, an integrated multicellular analysis in the microfluidic device is expected to improve accessibility, scalability, and practicality for end users.
Collapse
Affiliation(s)
- Chae Yun Bae
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jaejung Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Je-Kyun Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
44
|
|
45
|
Li N, Sun G, Wang S, Wang Y, Xiu Z, Sun D, Guo X, Zhang Y, Ma X. Engineering islet for improved performance by optimized reaggregation in alginate gel beads. Biotechnol Appl Biochem 2017; 64:400-405. [PMID: 26936645 DOI: 10.1002/bab.1489] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/28/2016] [Indexed: 12/22/2022]
Abstract
After islet isolation, diffusion has become the main mechanism to transport oxygen and nutrients into the core of islets. However, diffusion has limitations, by which nutrients cannot effectively reach the core of large islets and can eventually cause core cell death and islet loss. This problem can be resolved by dispersing islets into single islet cells, but single islet cells do not exhibit insulin release function in in vitro culture. In this study, we intended to establish a new islet engineering approach by forming islet cell clusters to improve islet survival and function. Therefore, alginate gels were used to encapsulate islet cells to form artificial islets after dispersion of islets into single cells. The shape of the islet cell clusters was similar to native islets, and the size of the islet cell clusters was limited to a maximum diameter of 100 μm. By limiting the diameter of this engineered islet cell cluster, cell viability was nearly 100%, a significant improvement over natural islets. Importantly, islet cell clusters express the genes of islets, including Isl-1, Gcg, and insulin-1, and insulin secretion ability was maintained in vitro.
Collapse
Affiliation(s)
- Na Li
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China.,School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Guangwei Sun
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Shujun Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Yu Wang
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zhilong Xiu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Dongsheng Sun
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China.,School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xin Guo
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Ying Zhang
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Xiaojun Ma
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| |
Collapse
|
46
|
Neuronal production from induced pluripotent stem cells in self-assembled collagen-hyaluronic acid-alginate microgel scaffolds with grafted GRGDSP/Ln5-P4. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:760-774. [PMID: 28482588 DOI: 10.1016/j.msec.2017.03.133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/22/2023]
Abstract
Self-assembled microgel functionalized with peptides was developed and applied to regenerate neurons from induced pluripotent stem cells (iPSCs). Collagen (COL), hyaluronic acid (HA), and alginate (ALG) were modified with methacrylic anhydride (MA), photocrosslinked for patterned particles, grafted with GRGDSP and Ln5-P4, and self-assembled to integrate the microgel into three-dimensional scaffolds. Physicochemical assessments revealed that the ternary microgel scaffolds had an optimal chemical composition at COLMA:HAMA:ALGMA=1:2:1. In fabricating cell-laden constructs, modified GRGDSP/Ln5-P4 in linear self-assembled scaffolds could significantly improve the entrapment efficiency and viability of iPSCs. In addition, GRGDSP/Ln5-P4 in the microgel constructs triggered the differentiation of iPSCs toward neurons, since the percentage of neurite-like cells could be higher than 98% after induction of nerve growth factor. Self-assembled microgel comprising COLMA, HAMA, ALGMA, and GRGDSP/Ln5-P4 may be promising in producing mature neural lineage from iPSCs, to provide better treatment for damaged nervous tissue.
Collapse
|
47
|
Ozawa F, Okitsu T, Takeuchi S. Improvement in the Mechanical Properties of Cell-Laden Hydrogel Microfibers Using Interpenetrating Polymer Networks. ACS Biomater Sci Eng 2017; 3:392-398. [DOI: 10.1021/acsbiomaterials.6b00619] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Fumisato Ozawa
- Institute
of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Takeuchi
Biohybrid Innovation Project, Exploratory Research for Advanced Technology
(ERATO), Japan Science and Technology (JST), Komaba Open Laboratory (KOL), Room
M202, 4-6-1, Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Teru Okitsu
- Institute
of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Takeuchi
Biohybrid Innovation Project, Exploratory Research for Advanced Technology
(ERATO), Japan Science and Technology (JST), Komaba Open Laboratory (KOL), Room
M202, 4-6-1, Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Shoji Takeuchi
- Institute
of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Takeuchi
Biohybrid Innovation Project, Exploratory Research for Advanced Technology
(ERATO), Japan Science and Technology (JST), Komaba Open Laboratory (KOL), Room
M202, 4-6-1, Komaba, Meguro-ku, Tokyo 153-8904, Japan
| |
Collapse
|
48
|
Wang K, Wang X, Han C, Hou W, Wang J, Chen L, Luo Y. From Micro to Macro: The Hierarchical Design in a Micropatterned Scaffold for Cell Assembling and Transplantation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28066985 DOI: 10.1002/adma.201604600] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 09/21/2016] [Indexed: 05/09/2023]
Abstract
A microwell-patterned membranous scaffold that integrates nano- and microscale topographical characteristics based on polyurethane is fabricated for transplanting syngeneic islets and allogeneic mesenchymal stem cells into diabetic rodents. The scaffold effectively allows for assembling of single cells/microtissues, enables the transplantation of cells with spatial control, and improves the transplant's engraftment efficacy in vivo for treating diabetes.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Xi Wang
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Chengsheng Han
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Wenda Hou
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Jinyang Wang
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Liangyi Chen
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Ying Luo
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
49
|
Abstract
Bioartificial pancreas made of insulin-secreting islets cells holds great promise in the treatment of individuals with Type-1 diabetes. Successful islet cell microencapsulation in biopolymers is a key step for providing immunoisolation of transplanted islet cells. Because of the variability in the size and shape of pancreatic islets, one of the main obstacles in their microencapsulation is the inability to consistently control shape, size, and microstructure of the encapsulating biopolymer capsule. In this chapter, we provide a detailed description of a microfluidic approach to islet cell encapsulation in alginate that might address the microencapsulation challenges.
Collapse
|
50
|
|