1
|
Jung YJ, Choi H, Choi IS, Lee JK. Biocompatible hydrogel coating on single living cells through visible light-induced polymerization. Chem Commun (Camb) 2024; 61:322-325. [PMID: 39632649 DOI: 10.1039/d4cc03075c] [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: 12/07/2024]
Abstract
Visible light-mediated photocatalysis leads to the efficient hydrogel coating of individual mammalian cells, functionalized with biocompatible anchor molecules tagged with fluorescein serving as a trifecta: photocatalyst, initiator, and fluorophore. NIH3T3 fibroblast cells are encapsulated within hydrogel shells of poly(ethylene glycol) diacrylate (PEGDA) and N-vinylpyrrolidone without any noticeable decrease in cell viability.
Collapse
Affiliation(s)
- Young Jae Jung
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, Daegu 41566, South Korea.
- Department of Chemistry, KAIST, Daejeon 34141, South Korea.
| | - Hyunwoo Choi
- Department of Chemistry, KAIST, Daejeon 34141, South Korea.
| | - Insung S Choi
- Department of Chemistry, KAIST, Daejeon 34141, South Korea.
| | - Jungkyu K Lee
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, Daegu 41566, South Korea.
| |
Collapse
|
2
|
Wang Q, Huang YX, Liu L, Zhao XH, Sun Y, Mao X, Li SW. Pancreatic islet transplantation: current advances and challenges. Front Immunol 2024; 15:1391504. [PMID: 38887292 PMCID: PMC11180903 DOI: 10.3389/fimmu.2024.1391504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Diabetes is a prevalent chronic disease that traditionally requires severe reliance on medication for treatment. Oral medication and exogenous insulin can only temporarily maintain blood glucose levels and do not cure the disease. Most patients need life-long injections of exogenous insulin. In recent years, advances in islet transplantation have significantly advanced the treatment of diabetes, allowing patients to discontinue exogenous insulin and avoid complications.Long-term follow-up results from recent reports on islet transplantation suggest that they provide significant therapeutic benefit although patients still require immunotherapy, suggesting the importance of future transplantation strategies. Although organ shortage remains the primary obstacle for the development of islet transplantation, new sources of islet cells, such as stem cells and porcine islet cells, have been proposed, and are gradually being incorporated into clinical research. Further research on new transplantation sites, such as the subcutaneous space and mesenteric fat, may eventually replace the traditional portal vein intra-islet cell infusion. Additionally, the immunological rejection reaction in islet transplantation will be resolved through the combined application of immunosuppressant agents, islet encapsulation technology, and the most promising mesenchymal stem cells/regulatory T cell and islet cell combined transplantation cell therapy. This review summarizes the progress achieved in islet transplantation, and discusses the research progress and potential solutions to the challenges faced.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yu-xi Huang
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao-hong Zhao
- Department of Pharmacy, Taizhou Hospital, Zhejiang University, Taizhou, Zhejiang, China
| | - Yi Sun
- MRL Global Medical Affairs, MSD China, Shanghai, China
| | - Xinli Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques and Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shao-wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques and Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
3
|
Kioulaphides S, García AJ. Encapsulation and immune protection for type 1 diabetes cell therapy. Adv Drug Deliv Rev 2024; 207:115205. [PMID: 38360355 PMCID: PMC10948298 DOI: 10.1016/j.addr.2024.115205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/20/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
Type 1 Diabetes (T1D) involves the autoimmune destruction of insulin-producing β-cells in the pancreas. Exogenous insulin injections are the current therapy but are user-dependent and cannot fully recapitulate physiological insulin secretion dynamics. Since the emergence of allogeneic cell therapy for T1D, the Edmonton Protocol has been the most promising immunosuppression protocol for cadaveric islet transplantation, but the lack of donor islets, poor cell engraftment, and required chronic immunosuppression have limited its application as a therapy for T1D. Encapsulation in biomaterials on the nano-, micro-, and macro-scale offers the potential to integrate islets with the host and protect them from immune responses. This method can be applied to different cell types, including cadaveric, porcine, and stem cell-derived islets, mitigating the issue of a lack of donor cells. This review covers progress in the efforts to integrate insulin-producing cells from multiple sources to T1D patients as a form of cell therapy.
Collapse
Affiliation(s)
- Sophia Kioulaphides
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
4
|
Rana MM, De la Hoz Siegler H. Evolution of Hybrid Hydrogels: Next-Generation Biomaterials for Drug Delivery and Tissue Engineering. Gels 2024; 10:216. [PMID: 38667635 PMCID: PMC11049329 DOI: 10.3390/gels10040216] [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: 02/28/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Hydrogels, being hydrophilic polymer networks capable of absorbing and retaining aqueous fluids, hold significant promise in biomedical applications owing to their high water content, permeability, and structural similarity to the extracellular matrix. Recent chemical advancements have bolstered their versatility, facilitating the integration of the molecules guiding cellular activities and enabling their controlled activation under time constraints. However, conventional synthetic hydrogels suffer from inherent weaknesses such as heterogeneity and network imperfections, which adversely affect their mechanical properties, diffusion rates, and biological activity. In response to these challenges, hybrid hydrogels have emerged, aiming to enhance their strength, drug release efficiency, and therapeutic effectiveness. These hybrid hydrogels, featuring improved formulations, are tailored for controlled drug release and tissue regeneration across both soft and hard tissues. The scientific community has increasingly recognized the versatile characteristics of hybrid hydrogels, particularly in the biomedical sector. This comprehensive review delves into recent advancements in hybrid hydrogel systems, covering the diverse types, modification strategies, and the integration of nano/microstructures. The discussion includes innovative fabrication techniques such as click reactions, 3D printing, and photopatterning alongside the elucidation of the release mechanisms of bioactive molecules. By addressing challenges, the review underscores diverse biomedical applications and envisages a promising future for hybrid hydrogels across various domains in the biomedical field.
Collapse
Affiliation(s)
- Md Mohosin Rana
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada;
- Centre for Blood Research, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hector De la Hoz Siegler
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
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
|
Xue Z, Mei D, Zhang L. Advances in single-cell nanoencapsulation and applications in diseases. J Microencapsul 2022; 39:481-494. [PMID: 35998209 DOI: 10.1080/02652048.2022.2111472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Single-cell nanoencapsulation is a method of coating the surface of single cell with nanomaterials. In the early 20th century, with the introduction of various types of organic or inorganic nano-polymer materials, the selection of cell types, and the functional modification of the outer coating, this technology has gradually matured. Typical preparation methods include interfacial polycondensation, complex condensation, spray drying, microdroplet ejection, and layer-by-layer (LbL) self-assembly. The LbL assembly technology utilises nanomaterials with opposite charges deposited on cells by strong interaction (electrostatic interaction) or weak interaction (hydrogen bonding, hydrophobic interaction), which drives compounds to spontaneously form films with complete structure, stable performance and unique functions on cells. According to the needs of the disease, choosing appropriate cell types and biocompatible and biodegradable nanomaterials could achieve the purpose of promoting cell proliferation, immune isolation, reducing phagocytosis of the reticuloendothelial system, prolonging the circulation time in vivo, and avoiding repeated administration. Therefore, encapsulated cells could be utilised in various biomedical fields, such as cell catalysis, biotherapy, vaccine manufacturing and antitumor therapy. This article reviews cell nanoencapsulation therapies for diseases, including the various cell sources used, nanoencapsulation technology and the latest advances in preclinical and clinical research.
Collapse
Affiliation(s)
- Ziyang Xue
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| | - Dan Mei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Ladeira B, Custodio C, Mano J. Core-Shell Microcapsules: Biofabrication and Potential Applications in Tissue Engineering and Regenerative Medicine. Biomater Sci 2022; 10:2122-2153. [DOI: 10.1039/d1bm01974k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The construction of biomaterial scaffolds that accurately recreate the architecture of living tissues in vitro is a major challenge in the field of tissue engineering and regenerative medicine. Core-shell microcapsules...
Collapse
|
8
|
Dubay R, Urban JN, Darling EM. Single-Cell Microgels for Diagnostics and Therapeutics. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2009946. [PMID: 36329867 PMCID: PMC9629779 DOI: 10.1002/adfm.202009946] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Indexed: 05/14/2023]
Abstract
Cell encapsulation within hydrogel droplets is transforming what is feasible in multiple fields of biomedical science such as tissue engineering and regenerative medicine, in vitro modeling, and cell-based therapies. Recent advances have allowed researchers to miniaturize material encapsulation complexes down to single-cell scales, where each complex, termed a single-cell microgel, contains only one cell surrounded by a hydrogel matrix while remaining <100 μm in size. With this achievement, studies requiring single-cell resolution are now possible, similar to those done using liquid droplet encapsulation. Of particular note, applications involving long-term in vitro cultures, modular bioinks, high-throughput screenings, and formation of 3D cellular microenvironments can be tuned independently to suit the needs of individual cells and experimental goals. In this progress report, an overview of established materials and techniques used to fabricate single-cell microgels, as well as insight into potential alternatives is provided. This focused review is concluded by discussing applications that have already benefited from single-cell microgel technologies, as well as prospective applications on the cusp of achieving important new capabilities.
Collapse
Affiliation(s)
- Ryan Dubay
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
- Draper, 555 Technology Sq., Cambridge, MA 02139, USA
| | - Joseph N Urban
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
| | - Eric M Darling
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Center for Biomedical Engineering, School of Engineering, Department of Orthopaedics, Brown University, 175 Meeting St., Providence, RI 02912, USA
| |
Collapse
|
9
|
Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater 2021; 133:87-101. [PMID: 34102338 PMCID: PMC9148663 DOI: 10.1016/j.actbio.2021.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
The effective suppression of adaptive immune responses is essential for the success of allogeneic cell therapies. In islet transplantation for Type 1 Diabetes, pre-existing autoimmunity provides an additional hurdle, as memory autoimmune T cells mediate both an autoantigen-specific attack on the donor beta cells and an alloantigen-specific attack on the donor graft cells. Immunosuppressive agents used for islet transplantation are generally successful in suppressing alloimmune responses, but dramatically hinder the widespread adoption of this therapeutic approach and fail to control memory T cell populations, which leaves the graft vulnerable to destruction. In this review, we highlight the capacity of biomaterials to provide local and nuanced instruction to suppress or alter immune pathways activated in response to an allogeneic islet transplant. Biomaterial immunoisolation is a common approach employed to block direct antigen recognition and downstream cell-mediated graft destruction; however, immunoisolation alone still permits shed donor antigens to escape into the host environment, resulting in indirect antigen recognition, immune cell activation, and the creation of a toxic graft site. Designing materials to decrease antigen escape, improve cell viability, and increase material compatibility are all approaches that can decrease the local release of antigen and danger signals into the implant microenvironment. Implant materials can be further enhanced through the local delivery of anti-inflammatory, suppressive, chemotactic, and/or tolerogenic agents, which serve to control both the innate and adaptive immune responses to the implant with a benefit of reduced systemic effects. Lessons learned from understanding how to manipulate allogeneic and autogenic immune responses to pancreatic islets can also be applied to other cell therapies to improve their efficacy and duration. STATEMENT OF SIGNIFICANCE: This review explores key immunologic concepts and critical pathways mediating graft rejection in Type 1 Diabetes, which can instruct the future purposeful design of immunomodulatory biomaterials for cell therapy. A summary of immunological pathways initiated following cellular implantation, as well as current systemic immunomodulatory agents used, is provided. We then outline the potential of biomaterials to modulate these responses. The capacity of polymeric encapsulation to block some powerful rejection pathways is covered. We also highlight the role of cellular health and biocompatibility in mitigating immune responses. Finally, we review the use of bioactive materials to proactively modulate local immune responses, focusing on key concepts of anti-inflammatory, suppressive, and tolerogenic agents.
Collapse
Affiliation(s)
- Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
10
|
Encapsulation Strategies for Pancreatic Islet Transplantation without Immune Suppression. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Kuwabara R, Hu S, Smink AM, Orive G, Lakey JRT, de Vos P. Applying Immunomodulation to Promote Longevity of Immunoisolated Pancreatic Islet Grafts. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:129-140. [PMID: 33397201 DOI: 10.1089/ten.teb.2020.0326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Islet transplantation is a promising therapy for insulin-dependent diabetes, but large-scale application is hampered by the lack of a consistent source of insulin-producing cells and need for lifelong administration of immunosuppressive drugs, which are associated with severe side effects. To avoid chronic immunosuppression, islet grafts can be enveloped in immunoisolating polymeric membranes. These immunoisolating polymeric membranes protect islet grafts from cell-mediated rejection while allowing diffusion of oxygen, nutrients, and insulin. Although clinical trials have shown the safety and feasibility of encapsulated islets to control glucose homeostasis, the strategy does up till now not support long-term graft survival. This partly can be explained by a significant loss of insulin-producing cells in the immediate period after implantation. The loss can be prevented by combining immunoisolation with immunomodulation, such as combined administration of immunomodulating cytokines or coencapsulation of immunomodulating cell types such as regulatory T cells, mesenchymal stem cells, or Sertoli cells. Also, administration of specific antibodies or apoptotic donor leucocytes is considered to create a tolerant microenvironment around immunoisolated grafts. In this review, we describe the outcomes and limitations of these approaches, as well as the recent progress in immunoisolating devices. Impact statement Immunoisolation by enveloping islets in semipermeable membranes allows for successful transplantation of islet grafts in the absence of chronic immunosuppression, but the duration of graft survival is still not permanent. The reasons for long-term final graft failure is not fully understood, but combining immunoisolation with immunomodulation of tissues or host immune system has been proposed to enhance the longevity of grafts. This article reviews the recent progress and challenges of immunoisolation, as well as the benefits and feasibility of combining encapsulation approaches with immunomodulation to promote longevity of encapsulated grafts.
Collapse
Affiliation(s)
- Rei Kuwabara
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuxian Hu
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Jonathan R T Lakey
- Department of Surgery and Biomedical Engineering, University of California Irvine, Irvine, California, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Yin Chin S, Cheung Poh Y, Kohler AC, Compton JT, Hsu LL, Lau KM, Kim S, Lee BW, Lee FY, Sia SK. Additive manufacturing of hydrogel-based materials for next-generation implantable medical devices. Sci Robot 2021; 2. [PMID: 31289767 DOI: 10.1126/scirobotics.aah6451] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Implantable microdevices often have static components rather than moving parts, and exhibit limited biocompatibility. This paper demonstrates a fast manufacturing method which can produce features in biocompatible materials down to tens of microns in scale, with intricate and composite patterns in each layer. By exploiting unique mechanical properties of hydrogels, we developed a "locking mechanism" for precise actuation and movement of freely moving parts, which can provide functions such as valves, manifolds, rotors, pumps, and delivery of payloads. Hydrogel components could be tuned within a wide range of mechanical and diffusive properties, and can be controlled after implantation without a sustained power supply. In a mouse model of osteosarcoma, triggering of release of doxorubicin from the device over ten days showed high treatment efficacy and low toxicity, at one-tenth of a standard systemic chemotherapy dose. Overall, this platform, called "iMEMS", enables development of biocompatible implantable microdevices with a wide range of intricate moving components that can be wirelessly controlled on demand, in a manner that solves issues of device powering and biocompatibility.
Collapse
Affiliation(s)
- Sau Yin Chin
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Yukkee Cheung Poh
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Anne-Céline Kohler
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Jocelyn T Compton
- Department of Orthopedic Surgery, Columbia University Medical Center, 622 West 168 Street, New York, New York 10032, USA
| | - Lauren L Hsu
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Kathryn M Lau
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Sohyun Kim
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Benjamin W Lee
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Francis Y Lee
- Department of Orthopedic Surgery, Columbia University Medical Center, 622 West 168 Street, New York, New York 10032, USA
| | - Samuel K Sia
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| |
Collapse
|
13
|
Eren TN, Kariksiz N, Demirci G, Tuncel D, Okte N, Yagci Acar H, Avci D. Irgacure 2959-functionalized poly(ethyleneimine)s as improved photoinitiators: enhanced water solubility, migration stability and visible-light operation. Polym Chem 2021. [DOI: 10.1039/d1py00298h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
I2959-functionalized PEI based PPIs with 7–9 times higher water solubility than I2959, visible light activation, dual-curing properties and migration stability were synthesized.
Collapse
Affiliation(s)
- Tugce Nur Eren
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
| | | | - Gozde Demirci
- Graduate School of Materials Science & Engineering
- Koc University
- Istanbul
- Turkey
| | - Duygu Tuncel
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
| | - Neren Okte
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
| | - Havva Yagci Acar
- Department of Chemistry
- Koc University
- Istanbul
- Turkey
- Surface Science & Technology Center (KUYTAM)
| | - Duygu Avci
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
| |
Collapse
|
14
|
Fuchs S, Ernst AU, Wang LH, Shariati K, Wang X, Liu Q, Ma M. Hydrogels in Emerging Technologies for Type 1 Diabetes. Chem Rev 2020; 121:11458-11526. [DOI: 10.1021/acs.chemrev.0c01062] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alexander U. Ernst
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Long-Hai Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Xi Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
15
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
16
|
Abstract
The microfluidics field is at a critical crossroads. The vast majority of microfluidic devices are presently manufactured using micromolding processes that work very well for a reduced set of biocompatible materials, but the time, cost, and design constraints of micromolding hinder the commercialization of many devices. As a result, the dissemination of microfluidic technology-and its impact on society-is in jeopardy. Digital manufacturing (DM) refers to a family of computer-centered processes that integrate digital three-dimensional (3D) designs, automated (additive or subtractive) fabrication, and device testing in order to increase fabrication efficiency. Importantly, DM enables the inexpensive realization of 3D designs that are impossible or very difficult to mold. The adoption of DM by microfluidic engineers has been slow, likely due to concerns over the resolution of the printers and the biocompatibility of the resins. In this article, we review and discuss the various printer types, resolution, biocompatibility issues, DM microfluidic designs, and the bright future ahead for this promising, fertile field.
Collapse
Affiliation(s)
- Arman Naderi
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA;
| | - Nirveek Bhattacharjee
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA;
| | - Albert Folch
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, USA;
| |
Collapse
|
17
|
Macdougall LJ, Anseth K. Bioerodible Hydrogels Based on Photopolymerized Poly(ethylene glycol)-co-poly(α-hydroxy acid) Diacrylate Macromers. Macromolecules 2020. [DOI: 10.1021/acs.macromol.0c00030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Facklam AL, Volpatti LR, Anderson DG. Biomaterials for Personalized Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1902005. [PMID: 31495970 DOI: 10.1002/adma.201902005] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/26/2019] [Indexed: 05/13/2023]
Abstract
Cell therapy has already had an important impact on healthcare and provided new treatments for previously intractable diseases. Notable examples include mesenchymal stem cells for tissue regeneration, islet transplantation for diabetes treatment, and T cell delivery for cancer immunotherapy. Biomaterials have the potential to extend the therapeutic impact of cell therapies by serving as carriers that provide 3D organization and support cell viability and function. With the growing emphasis on personalized medicine, cell therapies hold great potential for their ability to sense and respond to the biology of an individual patient. These therapies can be further personalized through the use of patient-specific cells or with precision biomaterials to guide cellular activity in response to the needs of each patient. Here, the role of biomaterials for applications in tissue regeneration, therapeutic protein delivery, and cancer immunotherapy is reviewed, with a focus on progress in engineering material properties and functionalities for personalized cell therapies.
Collapse
Affiliation(s)
- Amanda L Facklam
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Lisa R Volpatti
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
19
|
Burke G, Barron V, Geever T, Geever L, Devine DM, Higginbotham CL. Evaluation of the materials properties, stability and cell response of a range of PEGDMA hydrogels for tissue engineering applications. J Mech Behav Biomed Mater 2019; 99:1-10. [PMID: 31319331 DOI: 10.1016/j.jmbbm.2019.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 05/16/2019] [Accepted: 07/06/2019] [Indexed: 01/12/2023]
Abstract
The main aim of this study was to examine the stability of a range of polyethyleneglycol dimethacrylate (PEGDMA) hydrogels over a 28-day period in simulated physiological solution. Upon optimisation of the ultraviolet (UV) curing conditions, the PEGDMA hydrogels were prepared using four different monomer concentrations (25, 50, 75 and 100 wt% PEGDMA) in water and cross-linked by photopolymerisation. Initial results revealed a correlation between monomer concentration and swelling behaviour, where a decrease in swelling was observed with increase in monomer content. On storage in physiological solutions at 37 °C, a decrease in the weight remaining of the hydrogels and the pH of the solutions was observed over a 28-day period. Using scanning electron microscopy, the surface topography of the hydrogels appeared to get smoother and in parallel changes in hydrophilicty were observed, with the biggest changes observed for the higher monomer concentrations where water contact angle values were seen to increase toward 90°. However, the mechanical properties remained relatively unaffected and there was no adverse effect on cell metabolic activity observed for cells grown in the presence of PEGDMA samples or using elution methods. Looking at the combination of mechanical chemical and thermal properties shown these results are an important finding for scaffolds intended for tissue engineering applications, where provision of mechanical support without the elicitation of an inflammatory response due to polymer degradation products is crucial for successful integration and neotissue formation during the first 28 days post implantation.
Collapse
Affiliation(s)
- Gavin Burke
- Materials Research Institute, Athlone Institute of Technology, Dublin Road, Co. Westmeath, Ireland
| | - Valerie Barron
- Materials Research Institute, Athlone Institute of Technology, Dublin Road, Co. Westmeath, Ireland
| | - Tess Geever
- Materials Research Institute, Athlone Institute of Technology, Dublin Road, Co. Westmeath, Ireland
| | - Luke Geever
- Materials Research Institute, Athlone Institute of Technology, Dublin Road, Co. Westmeath, Ireland
| | - Declan M Devine
- Materials Research Institute, Athlone Institute of Technology, Dublin Road, Co. Westmeath, Ireland.
| | - Clement L Higginbotham
- Materials Research Institute, Athlone Institute of Technology, Dublin Road, Co. Westmeath, Ireland.
| |
Collapse
|
20
|
Hu S, de Vos P. Polymeric Approaches to Reduce Tissue Responses Against Devices Applied for Islet-Cell Encapsulation. Front Bioeng Biotechnol 2019; 7:134. [PMID: 31214587 PMCID: PMC6558039 DOI: 10.3389/fbioe.2019.00134] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Immunoisolation of pancreatic islets is a technology in which islets are encapsulated in semipermeable but immunoprotective polymeric membranes. The technology allows for successful transplantation of insulin-producing cells in the absence of immunosuppression. Different approaches of immunoisolation are currently under development. These approaches involve intravascular devices that are connected to the bloodstream and extravascular devices that can be distinguished in micro- and macrocapsules and are usually implanted in the peritoneal cavity or under the skin. The technology has been subject of intense fundamental research in the past decade. It has co-evolved with novel replenishable cell sources for cure of diseases such as Type 1 Diabetes Mellitus that need to be protected for the host immune system. Although the devices have shown significant success in animal models and even in human safety studies most technologies still suffer from undesired tissue responses in the host. Here we review the past and current approaches to modulate and reduce tissue responses against extravascular cell-containing micro- and macrocapsules with a focus on rational choices for polymer (combinations). Choices for polymers but also choices for crosslinking agents that induce more stable and biocompatible capsules are discussed. Combining beneficial properties of molecules in diblock polymers or application of these molecules or other anti-biofouling molecules have been reviewed. Emerging are also the principles of polymer brushes that prevent protein and cell-adhesion. Recently also immunomodulating biomaterials that bind to specific immune receptors have entered the field. Several natural and synthetic polymers and even combinations of these polymers have demonstrated significant improvement in outcomes of encapsulated grafts. Adequate polymeric surface properties have been shown to be essential but how the surface should be composed to avoid host responses remains to be identified. Current insight is that optimal biocompatible devices can be created which raises optimism that immunoisolating devices can be created that allows for long term survival of encapsulated replenishable insulin-producing cell sources for treatment of Type 1 Diabetes Mellitus.
Collapse
Affiliation(s)
- Shuixan Hu
- Division of Medical Biology, Department of Pathology and Medical Biology, Immunoendocrinology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | | |
Collapse
|
21
|
Davis KA, Wu PJ, Cahall CF, Li C, Gottipati A, Berron BJ. Coatings on mammalian cells: interfacing cells with their environment. J Biol Eng 2019; 13:5. [PMID: 30675178 PMCID: PMC6337841 DOI: 10.1186/s13036-018-0131-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/09/2018] [Indexed: 12/18/2022] Open
Abstract
The research community is intent on harnessing increasingly complex biological building blocks. At present, cells represent a highly functional component for integration into higher order systems. In this review, we discuss the current application space for cellular coating technologies and emphasize the relationship between the target application and coating design. We also discuss how the cell and the coating interact in common analytical techniques, and where caution must be exercised in the interpretation of results. Finally, we look ahead at emerging application areas that are ideal for innovation in cellular coatings. In all, cellular coatings leverage the machinery unique to specific cell types, and the opportunities derived from these hybrid assemblies have yet to be fully realized.
Collapse
Affiliation(s)
- Kara A. Davis
- Chemical and Materials Engineering, University of Kentucky, 177 FPAT, Lexington, KY 40506-0046 USA
| | - Pei-Jung Wu
- Chemical and Materials Engineering, University of Kentucky, 177 FPAT, Lexington, KY 40506-0046 USA
| | - Calvin F. Cahall
- Chemical and Materials Engineering, University of Kentucky, 177 FPAT, Lexington, KY 40506-0046 USA
| | - Cong Li
- Chemical and Materials Engineering, University of Kentucky, 177 FPAT, Lexington, KY 40506-0046 USA
| | - Anuhya Gottipati
- Chemical and Materials Engineering, University of Kentucky, 177 FPAT, Lexington, KY 40506-0046 USA
| | - Brad J. Berron
- Chemical and Materials Engineering, University of Kentucky, 177 FPAT, Lexington, KY 40506-0046 USA
| |
Collapse
|
22
|
Ernst AU, Bowers DT, Wang LH, Shariati K, Plesser MD, Brown NK, Mehrabyan T, Ma M. Nanotechnology in cell replacement therapies for type 1 diabetes. Adv Drug Deliv Rev 2019; 139:116-138. [PMID: 30716349 PMCID: PMC6677642 DOI: 10.1016/j.addr.2019.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/17/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
Islet transplantation is a promising long-term, compliance-free, complication-preventing treatment for type 1 diabetes. However, islet transplantation is currently limited to a narrow set of patients due to the shortage of donor islets and side effects from immunosuppression. Encapsulating cells in an immunoisolating membrane can allow for their transplantation without the need for immunosuppression. Alternatively, "open" systems may improve islet health and function by allowing vascular ingrowth at clinically attractive sites. Many processes that enable graft success in both approaches occur at the nanoscale level-in this review we thus consider nanotechnology in cell replacement therapies for type 1 diabetes. A variety of biomaterial-based strategies at the nanometer range have emerged to promote immune-isolation or modulation, proangiogenic, or insulinotropic effects. Additionally, coating islets with nano-thin polymer films has burgeoned as an islet protection modality. Materials approaches that utilize nanoscale features manipulate biology at the molecular scale, offering unique solutions to the enduring challenges of islet transplantation.
Collapse
Affiliation(s)
- Alexander U Ernst
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kaavian Shariati
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Mitchell D Plesser
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Natalie K Brown
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Tigran Mehrabyan
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
23
|
In situ photochemical crosslinking of hydrogel membrane for Guided Tissue Regeneration. Dent Mater 2018; 34:1769-1782. [PMID: 30336953 DOI: 10.1016/j.dental.2018.09.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Periodontitis is an inflammatory disease that destroys the tooth-supporting attachment apparatus. Guided tissue regeneration (GTR) is a technique based on a barrier membrane designed to prevent wound space colonization by gingival cells. This study examined a new formulation composed of two polymers that could be photochemically cross-linked in situ into an interpenetrated polymer network (IPN) forming a hydrogel membrane. METHODS We synthetized and characterized silanized hydroxypropyl methylcellulose (Si-HPMC) for its cell barrier properties and methacrylated carboxymethyl chitosan (MA-CMCS) for its degradable backbone to use in IPN. Hydrogel membranes were cross-linked using riboflavin photoinitiator and a dentistry visible light lamp. The biomaterial's physicochemical and mechanical properties were determined. Hydrogel membrane degradation was evaluated in lysozyme. Cytocompatibility was estimated by neutral red uptake. The cell barrier property was studied culturing human primary gingival fibroblasts or human gingival explants on membrane and analyzed with confocal microscopy and histological staining. RESULTS The IPN hydrogel membrane was obtained after 120s of irradiation. The IPN showed a synergistic increase in Young moduli compared with the single networks. The CMCS addition in IPN allows a progressive weight loss compared to each polymer network. Cytocompatibility was confirmed by neutral red assay. Human cell invasion was prevented by hydrogel membranes and histological sections revealed that the biomaterial exhibited a barrier effect in contact with soft gingival tissue. SIGNIFICANCE We demonstrated the ability of an innovative polymer formulation to form in situ, using a dentist's lamp, an IPN hydrogel membrane, which could be an easy-to-use biomaterial for GTR therapy.
Collapse
|
24
|
Chin SY, Poh YC, Kohler AC, Sia SK. An Additive Manufacturing Technique for the Facile and Rapid Fabrication of Hydrogel-based Micromachines with Magnetically Responsive Components. J Vis Exp 2018:56727. [PMID: 30080207 PMCID: PMC6126519 DOI: 10.3791/56727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Polyethylene glycol (PEG)-based hydrogels are biocompatible hydrogels that have been approved for use in humans by the FDA. Typical PEG-based hydrogels have simple monolithic architectures and often function as scaffolding materials for tissue engineering applications. More sophisticated structures typically take a long time to fabricate and do not contain moving components. This protocol describes a photolithography method that allows for facile and rapid microfabrication of PEG structures and devices. This strategy involves an in-house developed fabrication stage that allows for the rapid fabrication of 3D structures by building upwards in a layer-by-layer fashion. Independent moving components can also be aligned and assembled onto support structures to form integrated devices. These independent components are doped with superparamagnetic iron oxide nanoparticles that are sensitive to magnetic actuation. In this manner, the fabricated devices can be actuated using external magnets to yield movement of the components within. Hence, this technique allows for the fabrication of sophisticated MEMS-like devices (micromachines) that are composed entirely out of a biocompatible hydrogel, able to function without an onboard power source, and respond to a contact-less method of actuation. This manuscript describes the fabrication of both the fabrication set-up as well as the step-by-step method for the microfabrication of these hydrogels-based MEMS-like devices.
Collapse
Affiliation(s)
- Sau Yin Chin
- Molecular Engineering Laboratory, Biomedical Sciences Institute, Agency for Science Technology and Research; Department of Biomedical Engineering, Columbia University;
| | | | | | - Samuel K Sia
- Department of Biomedical Engineering, Columbia University
| |
Collapse
|
25
|
Pérez-Luna VH, González-Reynoso O. Encapsulation of Biological Agents in Hydrogels for Therapeutic Applications. Gels 2018; 4:E61. [PMID: 30674837 PMCID: PMC6209244 DOI: 10.3390/gels4030061] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 01/03/2023] Open
Abstract
Hydrogels are materials specially suited for encapsulation of biological elements. Their large water content provides an environment compatible with most biological molecules. Their crosslinked nature also provides an ideal material for the protection of encapsulated biological elements against degradation and/or immune recognition. This makes them attractive not only for controlled drug delivery of proteins, but they can also be used to encapsulate cells that can have therapeutic applications. Thus, hydrogels can be used to create systems that will deliver required therapies in a controlled manner by either encapsulation of proteins or even cells that produce molecules that will be released from these systems. Here, an overview of hydrogel encapsulation strategies of biological elements ranging from molecules to cells is discussed, with special emphasis on therapeutic applications.
Collapse
Affiliation(s)
- Víctor H Pérez-Luna
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, 10 West 33rd Street, Chicago, IL 60616, USA.
| | - Orfil González-Reynoso
- Departamento de Ingeniería Química, Universidad de Guadalajara, Blvd. Gral. Marcelino García Barragán # 1451, Guadalajara, Jalisco C.P. 44430, Mexico.
| |
Collapse
|
26
|
Takai M, Kawasaki Y, Arimoto S, Tanimoto Y, Kitamura Y, Sendo T. UV-irradiated 2-methyl-4'-(methylthio)-2-morpholinopropiophenone-containing injection solution produced frameshift mutations in the Ames mutagenicity assay. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:10135-10140. [PMID: 29488202 DOI: 10.1007/s11356-018-1539-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 02/13/2018] [Indexed: 06/08/2023]
Abstract
In previous studies, we detected the photoinitiators 1-hydroxycyclohexyl phenyl ketone (1-HCHPK), methyl 2-benzoylbenzoate (MBB), and 2-methyl-4'-(methylthio)-2-morpholinopropiophenone (MTMP) in intravenous injection solutions. In addition, we reported that 1-HCHPK, MBB, and MTMP exhibited cytotoxicity towards normal human peripheral blood mononuclear cells. A previous in vitro study reported that a free-radical photoinitiator introduced covalently bound purine residues into DNA. However, little is known about the in vitro mutagenicity of 1-HCHPK, MBB, and MTMP. In the present in vitro study, we evaluated the mutagenicity of 1-HCHPK, MBB, and MTMP using the Ames test. We found that untreated 1-HCHPK, MBB, and MTMP were not mutagenic in S. typhimurium strain TA97, TA98, TA100, TA102, or TA1535, regardless of the presence/absence of S9 activation. However, ultraviolet (UV) light-irradiated MTMP exhibited mutagenicity in S. typhimurium strain TA97 in the absence of S9 activation. In conclusion, we suggest that exposure to UV-irradiated MTMP, including in intravenous injection solutions, can result in frameshift mutations.
Collapse
Affiliation(s)
- Mariko Takai
- Department of Clinical Pharmacy, Dentistry, and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yoichi Kawasaki
- Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Sakae Arimoto
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka; Kita-ku, Okayama, 700-8530, Japan
| | - Yusuke Tanimoto
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka; Kita-ku, Okayama, 700-8530, Japan
| | - Yoshihisa Kitamura
- Department of Clinical Pharmacy, Dentistry, and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
- Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Toshiaki Sendo
- Department of Clinical Pharmacy, Dentistry, and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
- Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
27
|
Day JR, David A, Cichon AL, Kulkarni T, Cascalho M, Shikanov A. Immunoisolating poly(ethylene glycol) based capsules support ovarian tissue survival to restore endocrine function. J Biomed Mater Res A 2018; 106:1381-1389. [PMID: 29318744 DOI: 10.1002/jbm.a.36338] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 02/06/2023]
Abstract
A common irreversible adverse effect of life-saving anticancer treatments is loss of gonadal endocrine function and fertility, calling for a need to focus on post-treatment quality of life. Here, we investigated the use of poly(ethylene glycol)-vinyl sulfone (PEG-VS) based capsules to support syngeneic donor ovarian tissue for restoration of endocrine function in mice. We designed a dual immunoisolating capsule (PEG-Dual) by tuning the physical properties of the PEG hydrogels and combining proteolytically degradable and nondegradable layers to meet the numerous requirements for encapsulation and immunoisolation of ovarian tissue, such as nutrient diffusion and tissue expansion. Tuning the components of the PEG-Dual capsule to have similar physical properties allowed for concentric encapsulation. Upon implantation, the PEG-based capsules supported ovarian tissue survival and led to a significant decrease in follicle stimulating hormone levels 60 days postimplantation. Mice that received the implants resumed regular estrous cycle activity and follicle development in the implanted grafts. The PEG-Dual capsule provided an environment conducive for tissue survival, while providing a barrier to the host environment. This study demonstrated for the first time that immunoisolating PEG-VS capsules can support ovarian follicular development resulting in the restoration of ovarian endocrine function and can be applied to future allogeneic studies. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1381-1389, 2018.
Collapse
Affiliation(s)
- James R Day
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109
| | - Anu David
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109
| | - Alexa L Cichon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, 48109
| | - Tanay Kulkarni
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, 48109.,Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, 48109
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109.,Department of Macromolecular Science & Engineering, University of Michigan, Ann Arbor, Michigan, 48109
| |
Collapse
|
28
|
Day JR, David A, Kim J, Farkash EA, Cascalho M, Milašinović N, Shikanov A. The impact of functional groups of poly(ethylene glycol) macromers on the physical properties of photo-polymerized hydrogels and the local inflammatory response in the host. Acta Biomater 2018; 67:42-52. [PMID: 29242160 DOI: 10.1016/j.actbio.2017.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 01/03/2023]
Abstract
Poly(ethylene glycol) (PEG) can be functionalized and modified with various moieties allowing for a multitude of cross-linking chemistries. Here, we investigate how vinyl sulfone, acrylate, and maleimide functional end groups affect hydrogel formation, physical properties, viability of encapsulated cells, post-polymerization modification, and inflammatory response of the host. We have shown that PEG-VS hydrogels, in the presence of a co-monomer, N-vinyl-2-pyrrolidone (NVP), form more efficiently than PEG-Ac and PEG-Mal hydrogels, resulting in superior physical properties after 6 min of ultraviolet light exposure. PEG-VS hydrogels exhibited hydrolytic stability and non-fouling characteristics, as well as the ability to be modified with biological motifs, such as RGD, after polymerization. Additionally, unmodified PEG-VS hydrogels resulted in lesser inflammatory response, cellular infiltration, and macrophage recruitment after implantation for 28 days in mice. These findings show that altering the end group chemistry of PEG macromer impacts characteristics of the photo-polymerized network. We have developed a tunable non-degradable PEG system that is conducive for cell or tissue encapsulation and evokes a minimal inflammatory response, which could be utilized for future immunoisolation applications. STATEMENT OF SIGNIFICANCE The objective of this study was to develop a tunable non-degradable PEG system that is conducive for encapsulation and evokes a minimal inflammatory response, which could be utilized for immunoisolation applications. This study has demonstrated that reactive functional groups of the PEG macromers impact free radical mediated network formation. Here, we show PEG-VS hydrogels meet the design criteria for an immunoisolating device as PEG-VS hydrogels form efficiently via photo-polymerization, impacting bulk properties, was stable in physiological conditions, and elicited a minimal inflammatory response. Further, NVP can be added to the precursor solution to expedite the cross-linking process without impacting cellular response upon encapsulation. These findings present an additional approach/chemistry to encapsulate cells or tissue for immunoisolation applications.
Collapse
|
29
|
Abstract
Review of emerging advances and persisting challenges in the engineering and translation of islet encapsulation technologies.
Collapse
Affiliation(s)
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering
- Cornell University
- Ithaca
- USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering
- Cornell University
- Ithaca
- USA
| |
Collapse
|
30
|
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
|
31
|
Biocompatibility of hydrogel-based scaffolds for tissue engineering applications. Biotechnol Adv 2017; 35:530-544. [DOI: 10.1016/j.biotechadv.2017.05.006] [Citation(s) in RCA: 407] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 05/08/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
|
32
|
Alvarez-Urena P, Zhu B, Henslee G, Sonnet C, Davis E, Sevick-Muraca E, Davis A, Olmsted-Davis E. Development of a Cell-Based Gene Therapy Approach to Selectively Turn Off Bone Formation. J Cell Biochem 2017. [PMID: 28621436 DOI: 10.1002/jcb.26220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cell and gene therapy approaches are safer when they possess a system that enables the therapy to be rapidly halted. Human mesenchymal stem cells were transduced with an adenoviral vector containing the cDNA for bone morphogenetic protein 2 (AdBMP2) to induce bone formation. To make this method safer, a system to quickly kill these virally transduced cells was designed and evaluated. Cells were encapsulated inside poly(ethylene glycol) diacrylate (PEG-Da) hydrogels that are able to shield the cells from immunological destruction. The system involves an inducible caspase-9 (iCasp9) activated using a specific chemical inducer of dimerization (CID). Delivering AdBMP2-transduced human mesenchymal stem cells encapsulated in PEG-Da hydrogel promoted ectopic ossification in vivo, and the iCasp9 system allowed direct control of the timing of apoptosis of the injected cells. The iCasp9-CID system enhances the safety of delivering AdBMP2-transduced cells, making it a more compelling therapeutic for bone repair and spine fusion. J. Cell. Biochem. 118: 3627-3634, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pedro Alvarez-Urena
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Banghe Zhu
- Center for Molecular Imaging, University of Texas Health Sciences Center, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas
| | - Gabrielle Henslee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Corinne Sonnet
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eleanor Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eva Sevick-Muraca
- Center for Molecular Imaging, University of Texas Health Sciences Center, Houston, Texas
| | - Alan Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| | - Elizabeth Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
33
|
Poggioli R, Inverardi L, Ricordi C. Article Commentary: Islet Xenotransplantation. Cell Transplant 2017; 11:89-94. [DOI: 10.3727/096020198389816] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Raffaella Poggioli
- Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Ave., Miami, FL 33136
| | - Luca Inverardi
- Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Ave., Miami, FL 33136
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Ave., Miami, FL 33136
| |
Collapse
|
34
|
Miki A, Rivas-Carrillo JD, Navarro-Alvarez N, Soto-Gutierrez A, Chen Y, Tanaka K, Narushima M, Tabata Y, Okitsu T, Noguchi H, Matsumoto S, Tanaka N, Kobayashi N. Maintenance of Neovascularization at the Implantation Site of an Artificial Device by bFGF and Endothelial Cell Transplant. Cell Transplant 2017; 15:893-901. [PMID: 17299994 DOI: 10.3727/000000006783981378] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Development of a subcutaneously implantable bioartificial pancreas (BAP) with immunoisolatory function could have a great impact on the treatment of diabetes mellitus. We have developed an implantable BAP device with an ethylene vinyl alcohol (EVAL) membrane. In the present study, we used basic fibroblast growth factors (bFGF), which was incorporated in a carrier for sustained release, in order to induce neovascularization when the device was implanted subcutaneously. To maintain the vasculature thus formed, a cell infusion port was attached to the BAP device, through which the device was filled with human liver vascular endothelial cell line TMNK-1, and the vasculature could be adequately maintained. Mice were divided into the following three groups. In group 1, a bFGF-free BAP device was implanted subcutaneously. In group 2, a sustained-release bFGF-impregnated BAP device was implanted. In group 3, a sustained-release bFGF-impregnated BAP device was implanted, and 3 × 106 TMNK-1 cells were infused into the implanted device every week. Neovascularization induced in the subcutaneous tissue around the implanted BAP device was macroscopically examined and histologically evaluated. In addition, the tissue blood flow was measured using a laser blood flow meter. In mice in group 3, neovascularization was significantly induced and maintained until week 8 postimplantation. It was confirmed by scanning electron microscopy that infused TMNK-1 cells adhered to the inner polyethylene surface of the device. It was demonstrated that the use of bFGF and vascular endothelial TMNK-1 cells induced and maintained adequate vasculature and tissue blood flow surrounding the implantable bag-type BAP device. We believe that the present study will contribute to BAP development for the treatment of diabetes.
Collapse
Affiliation(s)
- Atsushi Miki
- Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Dwaine F. Emerich
- Department of Neuroscience, Alkermes, Inc, 64 Sidney Street, Cambridge MA 02139
| | - Heather C. Salzberg
- Department of Neuroscience, Alkermes, Inc, 64 Sidney Street, Cambridge MA 02139
| |
Collapse
|
36
|
Foster GA, García AJ. Bio-synthetic materials for immunomodulation of islet transplants. Adv Drug Deliv Rev 2017; 114:266-271. [PMID: 28532691 PMCID: PMC5581997 DOI: 10.1016/j.addr.2017.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/09/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022]
Abstract
Clinical islet transplantation is an effective therapy in restoring physiological glycemic control in type 1 diabetics. However, allogeneic islets derived from cadaveric sources elicit immune responses that result in acute and chronic islet destruction. To prevent immune destruction of islets, transplant recipients require lifelong delivery of immunosuppressive drugs, which are associated with debilitating side effects. Biomaterial-based strategies to eliminate the need for immunosuppressive drugs are an emerging therapy for improving islet transplantation. In this context, two main approaches have been used: 1) encapsulation of islets to prevent infiltration and contact of immune cells, and 2) local release of immunomodulatory molecules from biomaterial systems that suppress local immunity. Synthetic biomaterials provide excellent control over material properties, molecule presentation, and therapeutic release, and thus, are an emerging platform for immunomodulation to facilitate islet transplantation. This review highlights various synthetic biomaterial-based strategies for preventing immune rejection of islet allografts.
Collapse
Affiliation(s)
- Greg A Foster
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
37
|
Kamperman T, Henke S, van den Berg A, Shin SR, Tamayol A, Khademhosseini A, Karperien M, Leijten J. Single Cell Microgel Based Modular Bioinks for Uncoupled Cellular Micro- and Macroenvironments. Adv Healthc Mater 2017; 6. [PMID: 27973710 DOI: 10.1002/adhm.201600913] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/11/2016] [Indexed: 01/09/2023]
Abstract
Modular bioinks based on single cell microgels within distinct injectable prepolymers enable uncoupling of biomaterials' micro- and macroenvironments. These inks allow biofabrication of 3D constructs that recapitulate the multiscale modular design of native tissues with a single cell resolution. This approach represents a major step forward in endowing engineered constructs with the multifunctionality that underlies the behavior of native tissues.
Collapse
Affiliation(s)
- Tom Kamperman
- Department of Developmental BioEngineering; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Drienerlolaan 5 7500AE Enschede The Netherlands
| | - Sieger Henke
- Department of Developmental BioEngineering; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Drienerlolaan 5 7500AE Enschede The Netherlands
| | - Albert van den Berg
- BIOS Lab on a Chip group; MESA+ Institute for Nanotechnology; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; 7500AE Enschede The Netherlands
| | - Su Ryon Shin
- Biomaterials Innovation Research Center; Brigham and Women's Hospital; Harvard Medical School; 02139 Cambridge MA USA
- Harvard-MIT Division of Health Sciences and Technology; Massachusetts Institute of Technology; 02139 Cambridge MA USA
- Wyss Institute for Biologically Inspired Engineering; Harvard University; 02115 Boston MA USA
| | - Ali Tamayol
- Biomaterials Innovation Research Center; Brigham and Women's Hospital; Harvard Medical School; 02139 Cambridge MA USA
- Harvard-MIT Division of Health Sciences and Technology; Massachusetts Institute of Technology; 02139 Cambridge MA USA
- Wyss Institute for Biologically Inspired Engineering; Harvard University; 02115 Boston MA USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center; Brigham and Women's Hospital; Harvard Medical School; 02139 Cambridge MA USA
- Harvard-MIT Division of Health Sciences and Technology; Massachusetts Institute of Technology; 02139 Cambridge MA USA
- Wyss Institute for Biologically Inspired Engineering; Harvard University; 02115 Boston MA USA
- Department of Physics; King Abdulaziz University; 21589 Jeddah Saudi Arabia
- Department of Bioindustrial Technologies; College of Animal Bioscience and Technology; Konkuk University; Hwayang-dong, Gwangjin-gu Seoul 143-701 Republic of Korea
| | - Marcel Karperien
- Department of Developmental BioEngineering; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Drienerlolaan 5 7500AE Enschede The Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineering; MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente; Drienerlolaan 5 7500AE Enschede The Netherlands
- Biomaterials Innovation Research Center; Brigham and Women's Hospital; Harvard Medical School; 02139 Cambridge MA USA
- Harvard-MIT Division of Health Sciences and Technology; Massachusetts Institute of Technology; 02139 Cambridge MA USA
| |
Collapse
|
38
|
Alvarez-Urena P, Davis E, Sonnet C, Henslee G, Gugala Z, Strecker EV, Linscheid LJ, Cuchiara M, West J, Davis A, Olmsted-Davis E. Encapsulation of Adenovirus BMP2-Transduced Cells with PEGDA Hydrogels Allows Bone Formation in the Presence of Immune Response. Tissue Eng Part A 2017; 23:177-184. [PMID: 27967655 DOI: 10.1089/ten.tea.2016.0277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Gene therapy approaches have been difficult to implement due to pre-existing immunity against the virus used for delivery. To circumvent this problem, a cell-based approach was developed that avoided the use of free virus within the animal. However, even cells transduced in vitro with E1- to E3-deleted adenovirus encoding bone morphogenetic protein 2 (AdBMP2) resulted in the production of virus-neutralizing antibodies in mice. Furthermore, when mice received an intramuscular injection of nonencoding adenovirus (AdEmpty)-transduced cells, AdBMP2-transduced cells were unable to launch bone formation when an intramuscular injection of these BMP2-producing cells was delivered 1 week later. This phenomenon was not observed in NOD/SCID mice, and could be overcome in C57BL/6 mice by encapsulating the adenovirus-transduced cells in a nondegradable hydrogel poly(ethylene glycol) diacrylate (PEGDA). Data collectively suggest that PEGDA hydrogel encapsulation of AdBMP2-transduced cells prevents pre-existing immunity from suppressing BMP2-induced bone formation.
Collapse
Affiliation(s)
- Pedro Alvarez-Urena
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eleanor Davis
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Corinne Sonnet
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Gabrielle Henslee
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Zbigniew Gugala
- 2 Department of Orthopedic Surgery and Rehabilitation, The University of Texas Medical Branch at Galveston , Galveston, Texas
| | - Edward V Strecker
- 2 Department of Orthopedic Surgery and Rehabilitation, The University of Texas Medical Branch at Galveston , Galveston, Texas
| | - Laura J Linscheid
- 2 Department of Orthopedic Surgery and Rehabilitation, The University of Texas Medical Branch at Galveston , Galveston, Texas
| | - Maude Cuchiara
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Jennifer West
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Alan Davis
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,4 Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine , Houston, Texas.,5 Department of Orthopedic Surgery, Baylor College of Medicine , Houston, Texas
| | - Elizabeth Olmsted-Davis
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,4 Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine , Houston, Texas.,5 Department of Orthopedic Surgery, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
39
|
Cavallo A, Madaghiele M, Masullo U, Lionetto MG, Sannino A. Photo-crosslinked poly(ethylene glycol) diacrylate (PEGDA) hydrogels from low molecular weight prepolymer: Swelling and permeation studies. J Appl Polym Sci 2016. [DOI: 10.1002/app.44380] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Anna Cavallo
- Department of Engineering for Innovation; University of Salento; Lecce 73100 Italy
| | - Marta Madaghiele
- Department of Engineering for Innovation; University of Salento; Lecce 73100 Italy
| | - Ugo Masullo
- Department of Engineering for Innovation; University of Salento; Lecce 73100 Italy
| | - Maria Giulia Lionetto
- Department of Biological and Environmental Sciences and Technologies; University of Salento; Lecce 73100 Italy
| | - Alessandro Sannino
- Department of Engineering for Innovation; University of Salento; Lecce 73100 Italy
| |
Collapse
|
40
|
Lilly JL, Berron BJ. The Role of Surface Receptor Density in Surface-Initiated Polymerizations for Cancer Cell Isolation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:5681-9. [PMID: 27206735 PMCID: PMC5047530 DOI: 10.1021/acs.langmuir.6b01146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Fluid biopsies potentially offer a minimally invasive alternative to traditional tissue biopsies for the continual monitoring of metastatic cancer. Current established technologies for isolating circulating tumor cells (CTCs) suffer from poor purity and yield and require fixatives that preclude the collection of viable cells for longitudinal analyses of biological function. Antigen specific lysis (ASL) is a rapid, high-purity method of cell isolation based on targeted protective coatings on antigen-presenting cells and lysis depletion of unprotected antigen-negative cells. In ASL, photoinitiators are specifically labeled on cell surfaces that enable subsequent surface-initiated polymerization. Critically, the significant determinants of process yield have yet to be investigated for this emerging technology. In this work, we show that the labeling density of photoinitiators is strongly correlated with the yield of intact cells during ASL by flow cytometry analysis. Results suggest ASL is capable of delivering ∼25% of targeted cells after isolation using traditional antibody labeling approaches. Monomer formulations of two molecular weights of PEG-diacrylate (Mn ∼ 575 and 3500) are examined. The gelation response during ASL polymerization is also investigated via protein microarray analogues on planar glass. Finally, a density threshold of photoinitiator labeling required for protection during lysis is determined for both monomer formulations. These results indicate ASL is a promising technology for high yield CTC isolation for rare-cell function assays and fluid biopsies.
Collapse
Affiliation(s)
- Jacob L Lilly
- Department of Chemical and Materials Engineering, University of Kentucky , Lexington, Kentucky 40506, United States
| | - Brad J Berron
- Department of Chemical and Materials Engineering, University of Kentucky , Lexington, Kentucky 40506, United States
| |
Collapse
|
41
|
David A, Day J, Shikanov A. Immunoisolation to prevent tissue graft rejection: Current knowledge and future use. Exp Biol Med (Maywood) 2016; 241:955-61. [PMID: 27188513 DOI: 10.1177/1535370216647129] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This review focuses on the concept of immunoisolation and how this method has evolved over the last few decades. The concept of immunoisolation came out of the need to protect allogeneic transplant tissue from the host immune system and avoid systemic side effects of immunosuppression. The latter remains a significant hurdle in clinical translation of using tissue transplants for restoring endocrine function in diabetes, growth hormone deficiency, and other conditions. Herein, we review the most significant works studying the use of hydrogels, specifically alginate and poly (ethylene glycol), and membranes for immunoisolation and discuss how this approach can be applied in reproductive biology.
Collapse
Affiliation(s)
- Anu David
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - James Day
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
42
|
Huang S, Lee AJ, Tsoi R, Wu F, Zhang Y, Leong KW, You L. Coupling spatial segregation with synthetic circuits to control bacterial survival. Mol Syst Biol 2016; 12:859. [PMID: 26925805 PMCID: PMC4770385 DOI: 10.15252/msb.20156567] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Engineered bacteria have great potential for medical and environmental applications. Fulfilling this potential requires controllability over engineered behaviors and scalability of the engineered systems. Here, we present a platform technology, microbial swarmbot, which employs spatial arrangement to control the growth dynamics of engineered bacteria. As a proof of principle, we demonstrated a safeguard strategy to prevent unintended bacterial proliferation. In particular, we adopted several synthetic gene circuits to program collective survival in Escherichia coli: the engineered bacteria could only survive when present at sufficiently high population densities. When encapsulated by permeable membranes, these bacteria can sense the local environment and respond accordingly. The cells inside the microbial swarmbot capsules will survive due to their high densities. Those escaping from a capsule, however, will be killed due to a decrease in their densities. We demonstrate that this design concept is modular and readily generalizable. Our work lays the foundation for engineering integrated and programmable control of hybrid biological–material systems for diverse applications.
Collapse
Affiliation(s)
- Shuqiang Huang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Anna Jisu Lee
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ryan Tsoi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Feilun Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ying Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, NC, USA Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| |
Collapse
|
43
|
Combination strategy of multi-layered surface camouflage using hyperbranched polyethylene glycol and immunosuppressive drugs for the prevention of immune reactions against transplanted porcine islets. Biomaterials 2016; 84:144-156. [PMID: 26828680 DOI: 10.1016/j.biomaterials.2016.01.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 12/31/2015] [Accepted: 01/15/2016] [Indexed: 11/22/2022]
Abstract
This study suggests a novel method of stabilizing fragile porcine islets to prevent the dissociation after isolation and reducing immune cell invasion in a combination therapy of 'surface camouflaging' and immunosuppressive drugs (FK506, Rapamycin, MR-1, anti-CD19 mAb, and Clodrosome(®)) to effectively alleviate overall immune reactions against xenotransplanted porcine islets. The surface camouflage of pancreatic islets using biocompatible materials improved stabilization of pancreatic islet and prevented the infiltration of immune cells. Firstly, the surface of porcine islets was camouflaged by SH-6-arm-PEG-lipid and gelatin-catechol (artificial extracellular matrix) in order to stabilize the fragile isolated islets. Secondly, three different PEG layers (6-arm-PEG-SH, 6-arm-PEG-catechol, and linear PEG-SH) were chemically conjugated onto the surface of the stabilized porcine islets. Both artificial extracellular matrix (artificial ECM) and PEGylation effectively covered the surface of porcine islets without increasing the size of the whole islet. In addition, the viability and functionality of the islets were not affected by this multi-layer surface modification. The multi-layer modification significantly reduced the attachment of human serum albumin, fibronectin, and immunoglobulin G in comparison to the control collagen surface. The combination effect of multi-layer PEGylation and cocktailed immunosuppressive drugs on the survival time of the transplanted islets was assessed in a xenogeneic porcine-to-mouse model. The median survival time (MST) of 'artificial ECM + PEGylation' group was 4-fold increased compared to that of control group. In addition, the MST of 'artificial ECM + PEGylation + drug' group was 2.16-fold increased, compared to the 'control + drug' group. In conclusion, we proposed a novel porcine islet transplantation protocol using surface multi-layer modification and cocktailed immunosuppressive drugs, for stabilization and immunoprotection against xenogeneic immune reactions.
Collapse
|
44
|
Cell-laden Polymeric Microspheres for Biomedical Applications. Trends Biotechnol 2015; 33:653-666. [DOI: 10.1016/j.tibtech.2015.09.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/10/2015] [Accepted: 09/08/2015] [Indexed: 01/16/2023]
|
45
|
Zhu H, Yu L, He Y, Lyu Y, Wang B. Microencapsulated Pig Islet Xenotransplantation as an Alternative Treatment of Diabetes. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:474-89. [PMID: 26028249 DOI: 10.1089/ten.teb.2014.0499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Haitao Zhu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, China
- Heart Center, Northwest Women's and Children's Hospital, Xi'an, China
| | - Liang Yu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Yayi He
- Department of Endocrinology, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Yi Lyu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, China
- Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, China
- Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
46
|
Aijaz A, Faulknor R, Berthiaume F, Olabisi RM. Hydrogel Microencapsulated Insulin-Secreting Cells Increase Keratinocyte Migration, Epidermal Thickness, Collagen Fiber Density, and Wound Closure in a Diabetic Mouse Model of Wound Healing. Tissue Eng Part A 2015; 21:2723-32. [PMID: 26239745 PMCID: PMC4652158 DOI: 10.1089/ten.tea.2015.0069] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Wound healing is a hierarchical process of intracellular and intercellular signaling. Insulin is a potent chemoattractant and mitogen for cells involved in wound healing. Insulin's potential to promote keratinocyte growth and stimulate collagen synthesis in fibroblasts is well described. However, there currently lacks an appropriate delivery mechanism capable of consistently supplying a wound environment with insulin; current approaches require repeated applications of insulin, which increase the chances of infecting the wound. In this study, we present a novel cell-based therapy that delivers insulin to the wound area in a constant or glucose-dependent manner by encapsulating insulin-secreting cells in nonimmunogenic poly(ethylene glycol) diacrylate (PEGDA) hydrogel microspheres. We evaluated cell viability and insulin secretory characteristics of microencapsulated cells. Glucose stimulation studies verified free diffusion of glucose and insulin through the microspheres, while no statistical difference in insulin secretion was observed between cells in microspheres and cells in monolayers. Scratch assays demonstrated accelerated keratinocyte migration in vitro when treated with microencapsulated cells. In excisional wounds on the dorsa of diabetic mice, microencapsulated RIN-m cells accelerated wound closure by postoperative day 7; a statistically significant increase over AtT-20ins-treated and control groups. Histological results indicated significantly greater epidermal thickness in both microencapsulated RIN-m and AtT-20ins-treated wounds. The results suggest that microencapsulation enables insulin-secreting cells to persist long enough at the wound site for a therapeutic effect and thereby functions as an effective delivery vehicle to accelerate wound healing.
Collapse
Affiliation(s)
- Ayesha Aijaz
- Department of Biomedical Engineering, Rutgers University , Piscataway, New Jersey
| | - Renea Faulknor
- Department of Biomedical Engineering, Rutgers University , Piscataway, New Jersey
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University , Piscataway, New Jersey
| | - Ronke M Olabisi
- Department of Biomedical Engineering, Rutgers University , Piscataway, New Jersey
| |
Collapse
|
47
|
Grim JC, Marozas IA, Anseth KS. Thiol-ene and photo-cleavage chemistry for controlled presentation of biomolecules in hydrogels. J Control Release 2015; 219:95-106. [PMID: 26315818 DOI: 10.1016/j.jconrel.2015.08.040] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
Hydrogels have emerged as promising scaffolds in regenerative medicine for the delivery of biomolecules to promote healing. However, increasing evidence suggests that the context that biomolecules are presented to cells (e.g., as soluble verses tethered signals) can influence their bioactivity. A common approach to deliver biomolecules in hydrogels involves physically entrapping them within the network, such that they diffuse out over time to the surrounding tissues. While simple and versatile, the release profiles in such system are highly dependent on the molecular weight of the entrapped molecule relative to the network structure, and it can be difficult to control the release of two different signals at independent rates. In some cases, supraphysiologically high loadings are used to achieve therapeutic local concentrations, but uncontrolled release can then cause deleterious off-target side effects. In vivo, many growth factors and cytokines are stored in the extracellular matrix (ECM) and released on demand as needed during development, growth, and wound healing. Thus, emerging strategies in biomaterial chemistry have focused on ways to tether or sequester biological signals and engineer these bioactive scaffolds to signal to delivered cells or endogenous cells. While many strategies exist to achieve tethering of peptides, protein, and small molecules, this review focuses on photochemical methods, and their usefulness as a mild reaction that proceeds with fast kinetics in aqueous solutions and at physiological conditions. Photo-click and photo-caging methods are particularly useful because one can direct light to specific regions of the hydrogel to achieve spatial patterning. Recent methods have even demonstrated reversible introduction of biomolecules to mimic the dynamic changes of native ECM, enabling researchers to explore how the spatial and dynamic context of biomolecular signals influences important cell functions. This review will highlight how two photochemical methods have led to important advances in the tissue regeneration community, namely the thiol-ene photo-click reaction for bioconjugation and photocleavage reactions that allow for the removal of protecting groups. Specific examples will be highlighted where these methodologies have been used to engineer hydrogels that control and direct cell function with the aim of inspiring their use in regenerative medicine.
Collapse
Affiliation(s)
- Joseph C Grim
- Howard Hughes Medical Institute, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Ian A Marozas
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Kristi S Anseth
- Howard Hughes Medical Institute, University of Colorado at Boulder, Boulder, CO 80309, USA; Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
48
|
Ludwig B, Ludwig S. Transplantable bioartificial pancreas devices: current status and future prospects. Langenbecks Arch Surg 2015; 400:531-40. [DOI: 10.1007/s00423-015-1314-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 02/08/2023]
|
49
|
Kawasaki Y, Tsuboi C, Yagi K, Morizane M, Masaoka Y, Esumi S, Kitamura Y, Sendo T. Photoinitiators enhanced 1,2-dichloropropane-induced cytotoxicity in human normal embryonic lung fibroblasts cells in vitro. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:4763-4770. [PMID: 25501643 DOI: 10.1007/s11356-014-3939-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/01/2014] [Indexed: 06/04/2023]
Abstract
Dichloromethane (DCM) and 1,2-dichloropsropane (DCP) have various uses, including being solvents for paint removers. Photoinitiators are also used in a wide range of commercial applications such as printing. These chemicals have been shown to induce cytotoxic effects. In the present study, we evaluated the combined effects of DCM or DCP from paint removers and photoinitiators used in printing on normal human embryonic lung fibroblasts with the aim of preventing occupational injuries. We showed that DCP, 2,2-dimethoxy-2-phenylacetophenone (2,2-DMPAP), 2-ethylhexyl-4-(dimethylamino) benzoate (2-EHDAB), 1-hydroxycyclohexyl phenyl ketone (1-HCHPK), and methyl 2-benzoylbenzoate (MBB) induced cytotoxicity, whereas DCM and 2-isopropylthioxanthone (2-ITX) did not. In addition, 2-methyl-4'-(methylthio)-2-morpholinopropiophenone (MTMP) caused a slight increase in cytotoxicity. The combination of DCP and the four photoinitiators (2,2-DMPAP, 2-EHDAB, MBB, and MTMP) significantly induced cytotoxicity and also led to apoptosis. In conclusion, the combination of DCP and photoinitiators may increase the risk of respiratory diseases.
Collapse
Affiliation(s)
- Yoichi Kawasaki
- Department of Pharmacy, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Lin CC, Ki CS, Shih H. Thiol-norbornene photo-click hydrogels for tissue engineering applications. J Appl Polym Sci 2015; 132:41563. [PMID: 25558088 PMCID: PMC4280501 DOI: 10.1002/app.41563] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thiol-norbornene (thiol-ene) photo-click hydrogels have emerged as a diverse material system for tissue engineering applications. These hydrogels are cross-linked through light mediated orthogonal reactions between multi-functional norbornene-modified macromers (e.g., poly(ethylene glycol), hyaluronic acid, gelatin) and sulfhydryl-containing linkers (e.g., dithiothreitol, PEG-dithiol, bis-cysteine peptides) using low concentration of photoinitiator. The gelation of thiol-norbornene hydrogels can be initiated by long-wave UV light or visible light without additional co-initiator or co-monomer. The cross-linking and degradation behaviors of thiol-norbornene hydrogels are controlled through material selections, whereas the biophysical and biochemical properties of the gels are easily and independently tuned owing to the orthogonal reactivity between norbornene and thiol moieties. Uniquely, the cross-linking of step-growth thiol-norbornene hydrogels is not oxygen-inhibited, therefore the gelation is much faster and highly cytocompatible compared with chain-growth polymerized hydrogels using similar gelation conditions. These hydrogels have been prepared as tunable substrates for 2D cell culture, as microgels or bulk gels for affinity-based or protease-sensitive drug delivery, and as scaffolds for 3D cell culture. Reports from different laboratories have demonstrated the broad utility of thiol-norbornene hydrogels in tissue engineering and regenerative medicine applications, including valvular and vascular tissue engineering, liver and pancreas-related tissue engineering, neural regeneration, musculoskeletal (bone and cartilage) tissue regeneration, stem cell culture and differentiation, as well as cancer cell biology. This article provides an up-to-date overview on thiol-norbornene hydrogel cross-linking and degradation mechanisms, tunable material properties, as well as the use of thiol-norbornene hydrogels in drug delivery and tissue engineering applications.
Collapse
Affiliation(s)
- Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN. 46202, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN. 47907, USA
| | - Chang Seok Ki
- Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN. 46202, USA
- Department of Biosystems and Biomaterials Science and Engineering, Seoul National University, Seoul. 151-742 Republic of Korea
| | - Han Shih
- Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN. 46202, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN. 47907, USA
| |
Collapse
|