1
|
Bezold MG, Dollinger BR, DeJulius CR, Keech MC, Hanna AR, Kittel AR, Yu F, Gupta MK, D'Arcy R, Brunger JM, Duvall CL. Shear-thinning hydrogel for allograft cell transplantation and externally controlled transgene expression. Biomaterials 2025; 314:122812. [PMID: 39288619 DOI: 10.1016/j.biomaterials.2024.122812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
This work establishes the design of a fully synthetic, shear-thinning hydrogel platform that is injectable and can isolate engineered, allogeneic cell therapies from the host. We utilized RAFT to generate a library of linear random copolymers of N,N-dimethylacrylamide (DMA) and 2-vinyl-4,4-dimethyl azlactone (VDMA) with variable mol% VDMA and degree of polymerization. Poly(DMA-co-VDMA) copolymers were subsequently modified with either adamantane (Ad) or β-cyclodextrin (Cd) through amine-reactive VDMA to prepare hydrogel precursor macromers containing complementary guest-host pairing pendant groups that, when mixed, form shear-thinning hydrogels. Rheometric evaluation of the hydrogel library enabled identification of lead macromer structures comprising 15 mol% pendants (Ad or Cd) and a degree of polymerization of 1000; mixing of these Ad and Cd functionalized precursors yielded hydrogels possessing storage modulus above 1000 Pa, tan(δ) values below 1 and high yield strain, which are target characteristics of robust but injectable shear-thinning gels. This modular system proved amenable to nanoparticle integration with surface-modified nanoparticles displaying Ad. The addition of the Ad-functionalized nanoparticles simultaneously improved mechanical properties of the hydrogels and enabled extended hydrogel retention of a model small molecule in vivo. In studies benchmarking against alginate, a material traditionally used for cell encapsulation, the lead hydrogel showed significantly less fibrous encapsulation in a subcutaneous implant site. Finally, this platform was utilized to encapsulate and extend in vivo longevity of inducible transgene-engineered mesenchymal stem cells in an allogeneic transplant model. The hydrogels remained intact and blocked infiltration by host cells, consequently extending the longevity of grafted cell function relative to a benchmark, shear-thinning hyaluronic acid-based gel. In sum, the new synthetic, shear-thinning hydrogel system presented here shows potential for further development as an injectable platform for delivery and in situ drug modulation of allograft and engineered cell therapies.
Collapse
Affiliation(s)
- Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Bryan R Dollinger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Andrew R Hanna
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mukesh K Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
2
|
Costa L, Carvalho AF, Fernandes AJS, Campos T, Dourado N, Costa FM, Gama M. Bacterial nanocellulose as a simple and tailorable platform for controlled drug release. Int J Pharm 2024; 663:124560. [PMID: 39127171 DOI: 10.1016/j.ijpharm.2024.124560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
In this study we present a proof of concept of a simple and straightforward approach for the development of a Bacterial Nanocellulose drug delivery system (BNC-DDS), envisioning the local delivery of immunomodulatory drugs to prevent foreign body reaction (FBR). Inspired by the self-adhesion behavior of BNC upon drying, we proposed a BNC laminate entrapping commercial crystalline drugs (dexamethasone-DEX and GW2580) in a sandwich system. The stability of the bilayer BNC-DDS was evidenced by the high interfacial energy of the bilayer films, 150 ± 11 and 88 ± 7 J/m2 respectively for 2 mm- and 10-mm thick films, corresponding to an increase of 7.5 and 4.4-fold comparatively to commercial tissue adhesives. In vitro release experiments unveiled the tunability of the bilayer BNC-DDS by showing extended drug release when thicker BNC membranes were used (from 16 to 47 days and from 35 to 132 days, for the bilayer-BNC entrapping DEX and GW2580, respectively). Mathematical modeling of the release data pointed to a diffusion-driven mechanism with non-fickian behavior. Overall, the results have demonstrated the potential of this simple approach for developing BNC-drug depots for localized and sustained release of therapeutic agents over adjustable timeframes.
Collapse
Affiliation(s)
- Lígia Costa
- CEB - Centre of Biological Engineering, University of Minho, Campus Gualtar, Braga, Portugal; LABBELS -Associate Laboratory, Braga, Guimarães, Portugal
| | - Alexandre F Carvalho
- i3N and Physics Department, University of Aveiro Campus of Santiago, 3810-193 Aveiro, Portugal
| | - António J S Fernandes
- i3N and Physics Department, University of Aveiro Campus of Santiago, 3810-193 Aveiro, Portugal
| | - Teresa Campos
- LABBELS -Associate Laboratory, Braga, Guimarães, Portugal; CMEMS-UMINHO, Universidade do Minho, 4800-058 Guimarães, Portugal
| | - Nuno Dourado
- LABBELS -Associate Laboratory, Braga, Guimarães, Portugal; CMEMS-UMINHO, Universidade do Minho, 4800-058 Guimarães, Portugal
| | - Florinda M Costa
- i3N and Physics Department, University of Aveiro Campus of Santiago, 3810-193 Aveiro, Portugal
| | - Miguel Gama
- CEB - Centre of Biological Engineering, University of Minho, Campus Gualtar, Braga, Portugal; LABBELS -Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
3
|
Hou Y, Conklin B, Choi HK, Yang L, Lee KB. Probing Nanotopography-Mediated Macrophage Polarization via Integrated Machine Learning and Combinatorial Biophysical Cue Mapping. ACS NANO 2024; 18:25465-25477. [PMID: 39226301 DOI: 10.1021/acsnano.4c04406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Inflammatory responses, leading to fibrosis and potential host rejection, significantly hinder the long-term success and widespread adoption of biomedical implants. The ability to control and investigated macrophage inflammatory responses at the implant-macrophage interface would be critical for reducing chronic inflammation and improving tissue integration. Nonetheless, the systematic investigation of how surface topography affects macrophage polarization is typically complicated by the restricted complexity of accessible nanostructures, difficulties in achieving exact control, and biased preselection of experimental parameters. In response to these problems, we developed a large-scale, high-content combinatorial biophysical cue (CBC) array for enabling high-throughput screening (HTS) of the effects of nanotopography on macrophage polarization and subsequent inflammatory processes. Our CBC array, created utilizing the dynamic laser interference lithography (DLIL) technology, contains over 1 million nanotopographies, ranging from nanolines and nanogrids to intricate hierarchical structures with dimensions ranging from 100 nm to several microns. Using machine learning (ML) based on the Gaussian process regression algorithm, we successfully identified certain topographical signals that either repress (pro-M2) or stimulate (pro-M1) macrophage polarization. The upscaling of these nanotopographies for further examination has shown mechanisms such as cytoskeletal remodeling and ROCK-dependent epigenetic activation to be critical to the mechanotransduction pathways regulating macrophage fate. Thus, we have also developed a platform combining advanced DLIL nanofabrication techniques, HTS, ML-driven prediction of nanobio interactions, and mechanotransduction pathway evaluation. In short, our developed platform technology not only improves our ability to investigate and understand nanotopography-regulated macrophage inflammatory responses but also holds great potential for guiding the design of nanostructured coatings for therapeutic biomaterials and biomedical implants.
Collapse
Affiliation(s)
- Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai 200065, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
4
|
Zhou X, Cao W, Chen Y, Zhu Z, Lai Y, Liu Z, Jia F, Lu Z, Han H, Yao K, Wang Y, Ji J, Zhang P. An elastomer with in situ generated pure zwitterionic surfaces for fibrosis-resistant implants. Acta Biomater 2024; 185:226-239. [PMID: 38972625 DOI: 10.1016/j.actbio.2024.06.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/09/2024]
Abstract
Polymeric elastomers are widely utilized in implantable biomedical devices. Nevertheless, the implantation of these elastomers can provoke a robust foreign body response (FBR), leading to the rejection of foreign implants and consequently reducing their effectiveness in vivo. Building effective anti-FBR coatings on those implants remains challenging. Herein, we introduce a coating-free elastomer with superior immunocompatibility. A super-hydrophilic anti-fouling zwitterionic layer can be generated in situ on the surface of the elastomer through a simple chemical trigger. This elastomer can repel the adsorption of proteins, as well as the adhesion of cells, platelets, and diverse microbes. The elastomer elicited negligible inflammatory responses after subcutaneous implantation in rodents for 2 weeks. No apparent fibrotic capsule formation was observed surrounding the elastomer after 6 months in rodents. Continuous subcutaneous insulin infusion (CSII) catheters constructed from the elastomer demonstrated prolonged longevity and performance compared to commercial catheters, indicating its great potential for enhancing and extending the performance of various implantable biomedical devices by effectively attenuating local immune responses. STATEMENT OF SIGNIFICANCE: The foreign body response remains a significant challenge for implants. Complicated coating procedures are usually needed to construct anti-fibrotic coatings on implantable elastomers. Herein, a coating-free elastomer with superior immunocompatibility was achieved using a zwitterionic monomer derivative. A pure zwitterionic layer can be generated on the elastomer surface through a simple chemical trigger. This elastomer significantly reduces protein adsorption, cell and bacterial adhesion, and platelet activation, leading to minimal fibrotic capsule formation even after six months of subcutaneous implantation in rodents. CSII catheters constructed from the PQCBE-H elastomer demonstrated prolonged longevity and performance compared to commercial catheters, highlighting the significant potential of PQCBE-H elastomers for enhancing and extending the performance of various implantable biomedical devices.
Collapse
Affiliation(s)
- Xianchi Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China
| | - Wenzhong Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China
| | - Yongcheng Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China
| | - Zihao Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China
| | - Yuxian Lai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China
| | - Zuolong Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 311202, Zhejiang Province, PR China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, PR China
| | - Zhouyu Lu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, Zhejiang Province, PR China
| | - Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, Zhejiang Province, PR China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, Zhejiang Province, PR China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 311202, Zhejiang Province, PR China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang Province, PR China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 311202, Zhejiang Province, PR China.
| |
Collapse
|
5
|
Zhou X, Lu Z, Cao W, Zhu Z, Chen Y, Ni Y, Liu Z, Jia F, Ye Y, Han H, Yao K, Liu W, Wang Y, Ji J, Zhang P. Immunocompatible elastomer with increased resistance to the foreign body response. Nat Commun 2024; 15:7526. [PMID: 39214984 PMCID: PMC11364871 DOI: 10.1038/s41467-024-52023-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Polymeric elastomers are extensively employed to fabricate implantable medical devices. However, implantation of the elastomers can induce a strong immune rejection known as the foreign body response (FBR), diminishing their efficacy. Herein, we present a group of immunocompatible elastomers, termed easy-to-synthesize vinyl-based anti-FBR dense elastomers (EVADE). EVADE materials effectively suppress the inflammation and capsule formation in subcutaneous models of rodents and non-human primates for at least one year and two months, respectively. Implantation of EVADE materials significantly reduces the expression of inflammation-related proteins S100A8/A9 in adjacent tissues compared to polydimethylsiloxane. We also show that inhibition or knockout of S100A8/A9 leads to substantial attenuation of fibrosis in mice, suggesting a target for fibrosis inhibition. Continuous subcutaneous insulin infusion (CSII) catheters constructed from EVADE elastomers demonstrate significantly improved longevity and performance compared to commercial catheters. The EVADE materials reported here may enhance and extend function in various medical devices by resisting the local immune responses.
Collapse
Affiliation(s)
- Xianchi Zhou
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Zhouyu Lu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Zhejiang University, Hangzhou, P. R. China
| | - Wenzhong Cao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Zihao Zhu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Yifeng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Yanwen Ni
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Zuolong Liu
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Yang Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Zhejiang University, Hangzhou, P. R. China
| | - Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Zhejiang University, Hangzhou, P. R. China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Zhejiang University, Hangzhou, P. R. China
| | - Weifeng Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Jian Ji
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Peng Zhang
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| |
Collapse
|
6
|
Quiroz VM, Wang Y, Rakoski AI, Kasinathan D, Neshat SY, Hollister-Lock J, Doloff JC. Hydrogel Alginate Considerations for Improved 3D Matrix Stability and Cell Graft Viability and Function in Studying Type 1 Diabetes In Vitro. Adv Biol (Weinh) 2024; 8:e2300502. [PMID: 38243878 PMCID: PMC11259579 DOI: 10.1002/adbi.202300502] [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: 12/19/2023] [Indexed: 01/22/2024]
Abstract
Biomedical devices such as islet-encapsulating systems are used for treatment of type 1 diabetes (T1D). Despite recent strides in preventing biomaterial fibrosis, challenges remain for biomaterial scaffolds due to limitations on cells contained within. The study demonstrates that proliferation and function of insulinoma (INS-1) cells as well as pancreatic rat islets may be improved in alginate hydrogels with optimized gel%, crosslinking, and stiffness. Quantitative polymerase chain reaction (qPCR)-based graft phenotyping of encapsulated INS-1 cells and pancreatic islets identified a hydrogel stiffness range between 600 and 1000 Pa that improved insulin Ins and Pdx1 gene expression as well as glucose-sensitive insulin-secretion. Barium chloride (BaCl2) crosslinking time is also optimized due to toxicity of extended exposure. Despite possible benefits to cell viability, calcium chloride (CaCl2)-crosslinked hydrogels exhibited a sharp storage modulus loss in vitro. Despite improved stability, BaCl2-crosslinked hydrogels also exhibited stiffness losses over the same timeframe. It is believed that this is due to ion exchange with other species in culture media, as hydrogels incubated in dIH2O exhibited significantly improved stability. To maintain cell viability and function while increasing 3D matrix stability, a range of useful media:dIH2O dilution ratios for use are identified. Such findings have importance to carry out characterization and optimization of cell microphysiological systems with high fidelity in vitro.
Collapse
Affiliation(s)
- Victor M. Quiroz
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yuanjia Wang
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda I. Rakoski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Devi Kasinathan
- Department of Physiology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sarah Y. Neshat
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jennifer Hollister-Lock
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, Massachusetts 02215, USA
| | - Joshua C. Doloff
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
7
|
Zhou X, Wang Y, Ji J, Zhang P. Materials Strategies to Overcome the Foreign Body Response. Adv Healthc Mater 2024; 13:e2304478. [PMID: 38666550 DOI: 10.1002/adhm.202304478] [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: 12/15/2023] [Revised: 04/20/2024] [Indexed: 05/03/2024]
Abstract
The foreign body response (FBR) is an immune-mediated reaction that can occur with most biomaterials and biomedical devices. The FBR initiates a deterioration in the performance of implantable devices, representing a longstanding challenge that consistently hampers their optimal utilization. Over the last decade, significant strides are achieved based on either hydrogel design or surface modifications to mitigate the FBR. This review delves into recent material strategies aimed at mitigating the FBR. Further, the authors look forward to future novel anti-FBR materials from the perspective of clinical translation needs. Such prospective materials hold the potential to attenuate local immune responses, thereby significantly enhancing the overall performance of implantable devices.
Collapse
Affiliation(s)
- Xianchi Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 311202, P. R. China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 311202, P. R. China
| |
Collapse
|
8
|
Abebayehu D, Pfaff BN, Bingham GC, Miller AE, Kibet M, Ghatti S, Griffin DR, Barker TH. A Thy-1-negative immunofibroblast population emerges as a key determinant of fibrotic outcomes to biomaterials. SCIENCE ADVANCES 2024; 10:eadf2675. [PMID: 38875340 PMCID: PMC11177936 DOI: 10.1126/sciadv.adf2675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/10/2024] [Indexed: 06/16/2024]
Abstract
Fibrosis-associated fibroblasts have been identified across various fibrotic disorders, but not in the context of biomaterials, fibrotic encapsulation, and the foreign body response. In other fibrotic disorders, a fibroblast subpopulation defined by Thy-1 loss is strongly correlated with fibrosis yet we do not know what promotes Thy-1 loss. We have previously shown that Thy-1 is an integrin regulator enabling normal fibroblast mechanosensing, and here, leveraging nonfibrotic microporous annealed particle (MAP) hydrogels versus classical fibrotic bulk hydrogels, we demonstrate that Thy1-/- mice mount a fibrotic response to MAP gels that includes inflammatory signaling. We found that a distinct and cryptic α-smooth muscle actin-positive Thy-1- fibroblast population emerges in response to interleuklin-1β (IL-1β) and tumor necrosis factor-α (TNFα). Furthermore, IL-1β/TNFα-induced Thy-1- fibroblasts consist of two distinct subpopulations that are strongly proinflammatory. These findings illustrate the emergence of a unique proinflammatory, profibrotic fibroblast subpopulation that is central to fibrotic encapsulation of biomaterials.
Collapse
Affiliation(s)
- Daniel Abebayehu
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Robert Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Blaise N. Pfaff
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Grace C. Bingham
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Andrew E. Miller
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Mathew Kibet
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Surabhi Ghatti
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Donald R. Griffin
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Thomas H. Barker
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Robert Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
9
|
Wu J, Deng J, Theocharidis G, Sarrafian TL, Griffiths LG, Bronson RT, Veves A, Chen J, Yuk H, Zhao X. Adhesive anti-fibrotic interfaces on diverse organs. Nature 2024; 630:360-367. [PMID: 38778109 PMCID: PMC11168934 DOI: 10.1038/s41586-024-07426-9] [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: 03/25/2022] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
Implanted biomaterials and devices face compromised functionality and efficacy in the long term owing to foreign body reactions and subsequent formation of fibrous capsules at the implant-tissue interfaces1-4. Here we demonstrate that an adhesive implant-tissue interface can mitigate fibrous capsule formation in diverse animal models, including rats, mice, humanized mice and pigs, by reducing the level of infiltration of inflammatory cells into the adhesive implant-tissue interface compared to the non-adhesive implant-tissue interface. Histological analysis shows that the adhesive implant-tissue interface does not form observable fibrous capsules on diverse organs, including the abdominal wall, colon, stomach, lung and heart, over 12 weeks in vivo. In vitro protein adsorption, multiplex Luminex assays, quantitative PCR, immunofluorescence analysis and RNA sequencing are additionally carried out to validate the hypothesis. We further demonstrate long-term bidirectional electrical communication enabled by implantable electrodes with an adhesive interface over 12 weeks in a rat model in vivo. These findings may offer a promising strategy for long-term anti-fibrotic implant-tissue interfaces.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jue Deng
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Georgios Theocharidis
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Leigh G Griffiths
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Aristidis Veves
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hyunwoo Yuk
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- SanaHeal, Cambridge, MA, USA.
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
10
|
Xiang Y, Zhao Y, Cheng T, Sun S, Wang J, Pei R. Implantable Neural Microelectrodes: How to Reduce Immune Response. ACS Biomater Sci Eng 2024; 10:2762-2783. [PMID: 38591141 DOI: 10.1021/acsbiomaterials.4c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Implantable neural microelectrodes exhibit the great ability to accurately capture the electrophysiological signals from individual neurons with exceptional submillisecond precision, holding tremendous potential for advancing brain science research, as well as offering promising avenues for neurological disease therapy. Although significant advancements have been made in the channel and density of implantable neural microelectrodes, challenges persist in extending the stable recording duration of these microelectrodes. The enduring stability of implanted electrode signals is primarily influenced by the chronic immune response triggered by the slight movement of the electrode within the neural tissue. The intensity of this immune response increases with a higher bending stiffness of the electrode. This Review thoroughly analyzes the sequential reactions evoked by implanted electrodes in the brain and highlights strategies aimed at mitigating chronic immune responses. Minimizing immune response mainly includes designing the microelectrode structure, selecting flexible materials, surface modification, and controlling drug release. The purpose of this paper is to provide valuable references and ideas for reducing the immune response of implantable neural microelectrodes and stimulate their further exploration in the field of brain science.
Collapse
Affiliation(s)
- Ying Xiang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tingting Cheng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Shengkai Sun
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jine Wang
- Jiangxi Institute of Nanotechnology, Nanchang 330200, China
- College of Medicine and Nursing, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou 253023, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
11
|
Fathi P, Sundaresan V, Alfonso AL, Rama Varma A, Sadtler K. Factors Affecting the Evaluation of Collagen Deposition and Fibrosis In Vitro. Tissue Eng Part A 2024; 30:367-380. [PMID: 38511512 PMCID: PMC11250831 DOI: 10.1089/ten.tea.2023.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024] Open
Abstract
Immune responses to biomedical implants, wound healing, and diseased tissues often involve collagen deposition by fibroblasts and other stromal cells. Dysregulated collagen deposition can lead to complications, such as biomaterial fibrosis, cardiac fibrosis, desmoplasia, liver fibrosis, and pulmonary fibrosis, which can ultimately result in losses of organ function or failure of biomedical implants. Current in vitro methods to induce collagen deposition include growing the cells under macromolecular crowding conditions or on fibronectin-coated surfaces. However, the majority of these methods have been demonstrated with a single cell line, and the combined impacts of culture conditions and postculture processing on collagen deposition have not been explored in detail. In this work, the effects of macromolecular crowding versus fibronectin coating, fixation with methanol versus fixation with paraformaldehyde, and use of plastic substrates versus glass substrates were evaluated using the WI-38 human lung fibroblast cell line. Fibronectin coating was found to provide enhanced collagen deposition under macromolecular crowding conditions, while a higher plating density led to improved collagen I deposition compared with macromolecular crowding. Collagen deposition was found to be more apparent on plastic substrates than on glass substrates. The effects of primary cells versus cell lines, and mouse cells versus human cells, were evaluated using WI-38 cells, primary human lung fibroblasts, primary human dermal fibroblasts, primary mouse lung fibroblasts, primary mouse dermal fibroblasts, and the L929 mouse fibroblast cell line. Cell lines exhibited enhanced collagen I deposition compared with primary cells. Furthermore, collagen deposition was quantified with picrosirius red staining, and plate-based drug screening through picrosirius red staining of decellularized extracellular matrices was demonstrated. The results of this study provide detailed conditions under which collagen deposition can be induced in vitro in multiple cell types, with applications including material development, development of potential antifibrotic therapies, and mechanistic investigation of disease pathways. Impact Statement This study demonstrated the effects of cell type, biological conditions, fixative, culture substrate, and staining method on in vitro collagen deposition and visualization. Further the utility of plate-based picrosirius red staining of decellularized extracellular matrices for drug screening through collagen quantification was demonstrated. These results should provide clarity and a path forward for researchers who aim to conduct in vitro experiments on collagen deposition.
Collapse
Affiliation(s)
- Parinaz Fathi
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Unit for NanoEngineering and MicroPhysiological Systems (UNEMPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Vanathi Sundaresan
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Andrea Lucia Alfonso
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Anagha Rama Varma
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Unit for NanoEngineering and MicroPhysiological Systems (UNEMPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
12
|
Giri PS, Rath SN. Macrophage Polarization Dynamics in Biomaterials: Implications for in Vitro Wound Healing. ACS APPLIED BIO MATERIALS 2024; 7:2413-2422. [PMID: 38536097 DOI: 10.1021/acsabm.4c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The interaction between biomaterials and the immune system plays a pivotal role in determining the success or failure of implantable devices. Macrophages, as key orchestrators of immune responses, exhibit diverse reactions that influence tissue integration or lead to implant failure. This study focuses on unraveling the intricate relationship between macrophage phenotypes and biomaterials, specifically hydrogels, by employing THP-1 cells as a model. Through a comprehensive investigation using polysaccharide, polymer, and protein-based hydrogels, our research sheds light on how the properties of hydrogels influence macrophage polarization. Phenotypic observations, biochemical assays, surface marker expression, and gene expression profiles collectively demonstrate the differential macrophage polarization abilities of polysaccharide-, polymer-, and protein-based hydrogels. Moreover, our indirect coculture studies reveal that hydrogels fostering M2 polarization exhibit exceptional wound-healing capabilities. These findings highlight the crucial role of the hydrogel microenvironment in adjusting macrophage polarization, offering a fresh avenue for refining biomaterials to bolster advantageous immune responses and improve tissue integration. This research contributes valuable insights for designing biomaterials with tailored properties that can guide macrophage behavior, ultimately improving the overall success of implantable devices.
Collapse
Affiliation(s)
- Pravin Shankar Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284 Telangana, India
| | - Subha Narayan Rath
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284 Telangana, India
| |
Collapse
|
13
|
Zhang ZJ, Zhou Y, Tong H, Sun XC, Lv ZC, Yong JK, Wu YC, Xiang XL, Ding F, Zuo XL, Li F, Xia Q, Feng H, Fan CH. Programmable DNA Hydrogel Assisting Microcrystal Formulations for Sustained Locoregional Drug Delivery in Surgical Residual Tumor Lesions and Lymph Node Metastasis. Adv Healthc Mater 2024; 13:e2303762. [PMID: 38047767 DOI: 10.1002/adhm.202303762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Indexed: 12/05/2023]
Abstract
Surgical residual tumor lesions (R1 resection of surgical procedures (e.g., liver cancer infiltrating the diaphragm, surgical residual breast cancer, postoperative residual ovarian cancer) or boundary residual after ablation) and lymph node metastasis that cannot be surgically resected (retroperitoneal lymph nodes) significantly affect postoperative survival of tumor patients. This clinical conundrum poses three challenges for local drug delivery systems: stable and continuous delivery, good biocompatibility, and the ability to package new targeted drugs that can synergize with other treatments. Here, a drug-laden hydrogel generated from pure DNA strands and highly programmable in adjusting its mesh size is reported. Meanwhile, the DNA hydrogel can assist the microcrystallization of novel radiosensitizing drugs, ataxia telangiectasia and rad3-related protein (ATR) inhibitor (Elimusertib), further facilitating its long-term release. When applied to the tumor site, the hydrogel system demonstrates significant antitumor activity, minimized systemic toxicity, and has a modulatory effect on the tumor-immune cell interface. This drug-loaded DNA-hydrogel platform represents a novel modality for adjuvant therapy in patients with surgical residual tumor lesions and lymph node metastasis.
Collapse
Affiliation(s)
- Zi-Jie Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Shanghai Institute of Transplantation, Shanghai, 200127, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, 200127, China
| | - Yi Zhou
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - Huan Tong
- Shanghai First Maternity and Infant Hospital, Shanghai, 200127, China
| | - Xi-Cheng Sun
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - Zi-Cheng Lv
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - June-Kong Yong
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - Yi-Chi Wu
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - Xue-Lin Xiang
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - Fei Ding
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| | - Xiao-Lei Zuo
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, 200127, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Fan Li
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, 200127, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Shanghai Institute of Transplantation, Shanghai, 200127, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, 200127, China
| | - Hao Feng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Shanghai Institute of Transplantation, Shanghai, 200127, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, 200127, China
| | - Chun-Hai Fan
- Shanghai Institute of Transplantation, Shanghai, 200127, China
| |
Collapse
|
14
|
Zhou X, Hao H, Chen Y, Cao W, Zhu Z, Ni Y, Liu Z, Jia F, Wang Y, Ji J, Peng Zhang. Covalently grafted human serum albumin coating mitigates the foreign body response against silicone implants in mice. Bioact Mater 2024; 34:482-493. [PMID: 38292409 PMCID: PMC10827492 DOI: 10.1016/j.bioactmat.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Implantable biomaterials and biosensors are integral components of modern medical systems but often encounter hindrances due to the foreign body response (FBR). Herein, we report an albumin coating strategy aimed at addressing this challenge. Using a facile and scalable silane coupling strategy, human serum albumin (HSA) is covalently grafted to the surface of polydimethylsiloxane (PDMS) implants. This covalently grafted albumin coating remains stable and resistant to displacement by other proteins. Notably, the PDMS with covalently grafted HSA strongly resists the fibrotic capsule formation following a 180-day subcutaneous implantation in C57BL/6 mice. Furthermore, the albumin coating led to reduced recruitment of macrophages and triggered a mild immune activation pattern. Exploration of albumin coatings sourced from various mammalian species has shown that only HSA exhibited a promising anti-FBR effect. The albumin coating method reported here holds the potential to improve and extend the function of silicone-based implants by mitigating the host responses to subcutaneously implanted biomaterials.
Collapse
Affiliation(s)
- Xianchi Zhou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Hongye Hao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, PR China
| | - Yifeng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, PR China
| | - Wenzhong Cao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Zihao Zhu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Yanwen Ni
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Zuolong Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, PR China
- State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou, PR China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, PR China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, PR China
- State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
15
|
Lansberry TR, Stabler CL. Immunoprotection of cellular transplants for autoimmune type 1 diabetes through local drug delivery. Adv Drug Deliv Rev 2024; 206:115179. [PMID: 38286164 PMCID: PMC11140763 DOI: 10.1016/j.addr.2024.115179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune condition that results in the destruction of insulin-secreting β cells of the islets of Langerhans. Allogeneic islet transplantation could be a successful treatment for T1DM; however, it is limited by the need for effective, permanent immunosuppression to prevent graft rejection. Upon transplantation, islets are rejected through non-specific, alloantigen specific, and recurring autoimmune pathways. Immunosuppressive agents used for islet transplantation are generally successful in inhibiting alloantigen rejection, but they are suboptimal in hindering non-specific and autoimmune pathways. In this review, we summarize the challenges with cellular immunological rejection and therapeutics used for islet transplantation. We highlight agents that target these three immune rejection pathways and how to package them for controlled, local delivery via biomaterials. Exploring macro-, micro-, and nano-scale immunomodulatory biomaterial platforms, we summarize their advantages, challenges, and future directions. We hypothesize that understanding their key features will help identify effective platforms to prevent islet graft rejection. Outcomes can further be translated to other cellular therapies beyond T1DM.
Collapse
Affiliation(s)
- T R Lansberry
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - C L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Immunology and Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA.
| |
Collapse
|
16
|
Kim YW, Park JM, Park CS, Na H, Kang YW, Lee W, Sun JY. Anisotropically Conductive Hydrogels with Directionally Aligned PEDOT:PSS in a PVA Matrix. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4013-4023. [PMID: 38189267 DOI: 10.1021/acsami.3c16094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Electrical anisotropy, which is characterized by the efficient transmission of electrical signals in specific directions, is prevalent in both natural and engineered systems. However, traditional anisotropically conductive materials are often rigid and dry, thus limiting their utility in applications aiming for the seamless integration of various technologies with biological tissues. In the present study, we introduce a method for precisely controlling the microstructures of conductive and insulating polymers to create highly anisotropically conductive composite hydrogels. Our methodology involves combining aligned poly(vinyl alcohol) microfibrils, infused poly(3,4-ethylenedioxythiophene) polystyrenesulfonate, and sodium citrate precipitation to form dense, aligned conductive paths. This significantly enhances the electrical conductivity anisotropy (σ∥/σ⊥ ≈ 60.8) within these composite hydrogels.
Collapse
Affiliation(s)
- Yong-Woo Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Man Park
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea
| | - Chang Seo Park
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeonuk Na
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong-Woo Kang
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea
| | - Wooseop Lee
- Pohang Accelerator Laboratory, POSTECH, Pohang 37673, Republic of Korea
| | - Jeong-Yun Sun
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
17
|
Liu Z, Zhou X, Chen Y, Ni Y, Zhu Z, Cao W, Chen K, Yan Y, Ji J, Zhang P. Fibrous capsule-resistant, controllably degradable and functionalizable zwitterion-albumin hybrid hydrogels. Biomater Sci 2024; 12:468-478. [PMID: 38086632 DOI: 10.1039/d3bm01783d] [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: 01/17/2024]
Abstract
Foreign body response (FBR) represents an immune-mediated cascade reaction capable of inducing the rejection of foreign implants, thereby compromising their in vivo performance. Pure zwitterionic hydrogels have demonstrated the ability to resist long-term FBR, owing to their outstanding antifouling capabilities. However, achieving such a robust anti-FBR effect necessitates stringent requirements concerning the purity of zwitterionic materials, which constrains their broader functional applications. Herein, we present a biocompatible, controllably degradable, and functionalizable zwitterion-albumin hybrid hydrogel. The zwitterionic hydrogel crosslinked with serum albumin exhibits controllable degradation and excels in preventing the adsorption of various proteins and adhesion of cells and bacteria. Moreover, the hydrogel significantly alleviates the host's FBR compared with PEG hydrogels and particularly outperforms PEG-based cross-linker crosslinked zwitterionic hydrogels in reducing collagen encapsulation when subcutaneously implanted into mice. The zwitterion-albumin hybrid hydrogel shows potential as a functionalizable anti-FBR material in the context of implantable materials and biomedical devices.
Collapse
Affiliation(s)
- Zuolong Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Xianchi Zhou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yanwen Ni
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Zihao Zhu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Wenzhong Cao
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Kexin Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Yu Yan
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, P. R. China
| | - Peng Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, P. R. China.
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining, P. R. China
| |
Collapse
|
18
|
French A, Hollister-Lock J, Sullivan BA, Stas E, Hwa AJ, Weir GC, Bonner-Weir S. Enhancement of Subcutaneous Islet Transplant Performance by Collagen 1 Gel. Cell Transplant 2024; 33:9636897241283728. [PMID: 39361612 PMCID: PMC11457190 DOI: 10.1177/09636897241283728] [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: 05/20/2024] [Revised: 07/14/2024] [Accepted: 08/14/2024] [Indexed: 10/05/2024] Open
Abstract
Human islets can be transplanted into the portal vein for T1 diabetes, and a similar procedure is being used in a clinical trial for stem cell-derived beta-like cells. Efforts have been underway to find an alternative transplant site that will foster better islet cell survival and function. Although conceptually attractive, the subcutaneous (SC) site has yielded disappointing results, in spite of some improvements resulting from more attention paid to vascularization and differentiation factors, including collagen. We developed a method to transplant rat islets in a disk of type 1 collagen gel and found improved efficacy of these transplants. Survival of islets following transplantation (tx) was determined by comparing insulin content of the graft to that of the pre-transplant islets from the same isolation. At 14 days after transplantation, grafts of the disks had more than double the recovered insulin than islets transplanted in ungelled collagen. SC grafts of disks had similar insulin content to grafts in a kidney site and in epididymal fat pads. In vivo disks underwent contraction to 10% of initial volume within 24 h but the islets remained healthy and well distributed. Whole mount imaging showed that residual donor vascular cells within the islets expanded and connected to ingrowing host blood vessels. Islets (400 rat islet equivalents (IEQ)) in the collagen disks transplanted into an SC site of NOD scid IL2R gammanull (NSG) mice reversed streptozotocin (STZ)-induced diabetes within 10 days as effectively as transplants in the kidney site. Thus, a simple change of placing islets into a gel of collagen 1 prior to transplantation allowed a prompt reversal of STZ-induced diabetes using SC site.
Collapse
Affiliation(s)
- Anna French
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Jennifer Hollister-Lock
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Brooke A. Sullivan
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Eline Stas
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Albert J. Hwa
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Gordon C. Weir
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Susan Bonner-Weir
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| |
Collapse
|
19
|
Huang X, Li L, Ou C, Shen M, Li X, Zhang M, Wu R, Kou X, Gao L, Liu F, Luo R, Wu Q, Gong C. Tumor Environment Regression Therapy Implemented by Switchable Prune-to-Essence Nanoplatform Unleashed Systemic Immune Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303715. [PMID: 37875395 PMCID: PMC10724435 DOI: 10.1002/advs.202303715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/01/2023] [Indexed: 10/26/2023]
Abstract
Coevolution of tumor cells and surrounding stroma results in protective protumoral environment, in which abundant vessel, stiff structure and immunosuppression promote each other, cooperatively incurring deterioration and treatment compromise. Reversing suchenvironment may transform tumors from treatment-resistant to treatment-vulnerable. However, effective reversion requires synergistic comprehensive regression of such environment under precise control. Here, the first attempt to collaboratively retrograde coevolutionary tumor environment to pre-oncogenesis status, defined as tumor environment regression therapy, is made for vigorous immune response eruption by a switchable prune-to-essence nanoplatform (Pres) with simplified composition and fabrication process. Through magnetic targeting and multimodal imaging of Pres, tumor environment regression therapy is guided, optimized and accomplished in a trinity way: Antiangiogenesis is executed to rarefy vessels to impede tumor progression. By seizing the time, cancer associated fibroblasts are eliminated to diminish collagen and loosen the stiff structure for deep penetration of Pres, which alternately functioned in deeper tumors, forming a positive feedback loop. Through this loop, immune cell infiltration, immunosuppression mitigation and immunogenic cells death induction are all fulfilled and further escalated in the regressed environment. These transformations consequently unleashed systemic immune responses and generated immune memory against carcinoma. This study provides new insights intotreatment of solid tumors.
Collapse
Affiliation(s)
- Xianzhou Huang
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Lu Li
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Chunqing Ou
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Meiling Shen
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xinchao Li
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Miaomiao Zhang
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Rui Wu
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaorong Kou
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Ling Gao
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Furong Liu
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Rui Luo
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Qinjie Wu
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Changyang Gong
- Department of BiotherapyCancer center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
20
|
Huang R, Zhou P, Chen B, Zhu Y, Chen X, Min Y. Stimuli-Responsive Nanoadjuvant Rejuvenates Robust Immune Responses to Sensitize Cancer Immunotherapy. ACS NANO 2023; 17:21455-21469. [PMID: 37897704 DOI: 10.1021/acsnano.3c06233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2023]
Abstract
Despite their immense therapeutic potential, cancer immunotherapies such as immune checkpoint blockers (ICBs) benefit only a small subset of patients. Toll-like receptor agonists reverse the immunosuppressive tumor microenvironment (TME) to enhance antitumor immunity, but their systemic administration induces side effects. This work describes a TME-responsive nanotherapeutic platform for the site-specific release of drug candidates in tumors with a significant antitumor efficacy. Imidazoquinoline (IMQ)-derived liposomal nanovesicles (LN-IMQ) triggered the antitumor ability of macrophages, mobilized T-cell immunity, and promoted the secretion of antitumor cytokines, explaining the synergistic effect of LN-IMQ with ICBs. LN-IMQ monotherapy observed complete tumor regression in 6/8 of 4T1-bearing mouse, and cured mice resisted secondary tumor challenge. Besides, LN-IMQ decreased the occurrence of lung metastases, being effective against advanced metastases. On the other hand, neoantigen-based cancer vaccine has very low immune responses. Here, we also verified that LN-IMQ can serve as an ideal tumor antigen delivery vector. Cancer cells in vitro treated with chemotherapeutic drugs included multiple neoantigens and high levels of damage-associated molecular patterns, which were then successfully encapsulated in LN-IMQ to obtain a "personalized nanovaccine" with artificially amplified antigenicity and adjuvant properties. This study developed an attractive potential personalized nanovaccine for chemotherapeutic-drug-induced tumor neoantigens and immunotherapy.
Collapse
Affiliation(s)
- Ruijie Huang
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Peijie Zhou
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Bo Chen
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Yuanzeng Min
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
21
|
Fu M, Yang C, Sun G. Recent advances in immunomodulatory hydrogels biomaterials for bone tissue regeneration. Mol Immunol 2023; 163:48-62. [PMID: 37742359 DOI: 10.1016/j.molimm.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/27/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
There is a high incidence of fractures in clinical practice and therapy. The repairment of critical size defects in the skeletal system remains a huge challenge for surgeons and researchers, which can be overcame by the application of bone tissue-engineered biomaterials. An increasing number of investigations have revealed that the immune system plays a vital role in the repair of bone defects, especially macrophages, which can modulate the integration of biomaterials and bone regeneration in multiple ways. Therefore, it has become increasingly important in regenerative medicine to regulate macrophage polarization to prevent inflammation caused by biomaterial implantation. Recent studies have stressed the importance of hydrogel-based modifications and the incorporation of various cellular and molecular signals for regulating immune responses to promote bone tissue regeneration and integrate biomaterials. In this review, we first elaborate briefly on the described the general physiological mechanism and process of bone tissue regeneration. Then, we summarized the immunomodulatory role macrophages play in bone repair. In addition, the role of hydrogel-based immune modification targeting macrophage modulation in accelerating and enhancing bone tissue regeneration was also discussed. Finally, we highlighted future directions and research strategies related to hydrogel optimization for the regulation of the immune response during bone regeneration and healing.
Collapse
Affiliation(s)
- Mei Fu
- Guixin Sun - Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chensong Yang
- Guixin Sun - Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guixin Sun
- Guixin Sun - Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
22
|
Peng Z, Xie C, Jin S, Hu J, Yao X, Ye J, Zhang X, Lim JX, Wu B, Wu H, Liang R, Wen Y, Huang J, Zou X, Ouyang H. Biomaterial based implants caused remote liver fatty deposition through activated blood-derived macrophages. Biomaterials 2023; 301:122234. [PMID: 37421671 DOI: 10.1016/j.biomaterials.2023.122234] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023]
Abstract
Understanding the biocompatibility of biomaterials is a prerequisite for the prediction of its clinical application, and the present assessments mainly rely on in vitro cell culture and in situ histopathology. However, remote organs responses after biomaterials implantation is unclear. Here, by leveraging body-wide-transcriptomics data, we performed in-depth systems analysis of biomaterials - remote organs crosstalk after abdominal implantation of polypropylene and silk fibroin using a rodent model, demonstrating local implantation caused remote organs responses dominated by acute-phase responses, immune system responses and lipid metabolism disorders. Of note, liver function was specially disturbed, defined as hepatic lipid deposition. Combining flow cytometry analyses and liver monocyte recruitment inhibition experiments, we proved that blood derived monocyte-derived macrophages in the liver underlying the mechanism of abnormal lipid deposition induced by local biomaterials implantation. Moreover, from the perspective of temporality, the remote organs responses and liver lipid deposition of silk fibroin group faded away with biomaterial degradation and restored to normal at end, which highlighted its superiority of degradability. These findings were further indirectly evidenced by human blood biochemical ALT and AST examination from 141 clinical cases of hernia repair using silk fibroin mesh and polypropylene mesh. In conclusion, this study provided new insights on the crosstalk between local biomaterial implants and remote organs, which is of help for future selecting and evaluating biomaterial implants with the consideration of whole-body response.
Collapse
Affiliation(s)
- Zhi Peng
- Central Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shucheng Jin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiajie Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xudong Yao
- The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jinchun Ye
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xianzhu Zhang
- Central Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia Xuan Lim
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, China
| | - Bingbing Wu
- The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Haoyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ya Wen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiahui Huang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaohui Zou
- Central Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China.
| |
Collapse
|
23
|
Lee S, Park S, Park J, Lee JY. Implantable polypyrrole bioelectrodes inducing anti-inflammatory macrophage polarization for long-term in vivo signal recording. Acta Biomater 2023; 168:458-469. [PMID: 37414115 DOI: 10.1016/j.actbio.2023.06.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/06/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
Bioelectrodes are critical components of implantable electronic devices that enable precise electrical signal transmission in close contact with living tissues. However, their in vivo performance is often compromised by inflammatory tissue reactions mainly induced by macrophages. Hence, we aimed to develop implantable bioelectrodes with high performance and high biocompatibility by actively modulating the inflammatory response of macrophages. Consequently, we fabricated heparin-doped polypyrrole electrodes (PPy/Hep) and immobilized anti-inflammatory cytokines (interleukin-4 [IL-4]) via non-covalent interactions. IL-4 immobilization did not alter the electrochemical performance of the original PPy/Hep electrodes. In vitro primary macrophage culture revealed that IL-4-immobilized PPy/Hep electrodes induced anti-inflammatory polarization of macrophages, similar to the soluble IL-4 control. In vivo subcutaneous implantation indicated that IL-4 immobilization on PPy/Hep promoted the anti-inflammatory polarization of host macrophages and significantly mitigated scarring around the implanted electrodes. In addition, high-sensitivity electrocardiogram signals were recorded from the implanted IL-4-immobilized PPy/Hep electrodes and compared to bare gold and PPy/Hep electrodes, which were maintained for up to 15 days post-implantation. This simple and effective surface modification strategy for developing immune-compatible bioelectrodes will facilitate the development of various electronic medical devices that require high sensitivities and long-term stabilities. STATEMENT OF SIGNIFICANCE: To fabricate highly immunocompatible conductive polymer-based implantable electrodes with high performance and stability in vivo, we introduced the anti-inflammatory activity to PPy/Hep electrodes by immobilizing IL-4 via non-covalent surface modification. IL-4-immobilized PPy/Hep could significantly mitigate inflammatory responses and scarring around implants by skewing macrophages to an anti-inflammatory phenotype. The IL-4-immobilized PPy/Hep electrodes could successfully record in vivo electrocardiogram signals for up to 15 days with no substantial sensitivity loss, retaining their superior sensitivity compared to bare gold and pristine PPy/Hep electrodes. Our simple and effective surface modification strategy for developing immune-compatible bioelectrodes will facilitate the development of various electronic medical devices that require high sensitivities and long-term stabilities, such as neural electrode arrays, biosensors, and cochlear electrodes.
Collapse
Affiliation(s)
- Sanghun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Sehyeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
24
|
Wang X, Zhou X, Kang L, Lai Y, Ye H. Engineering natural molecule-triggered genetic control systems for tunable gene- and cell-based therapies. Synth Syst Biotechnol 2023; 8:416-426. [PMID: 37384125 PMCID: PMC10293594 DOI: 10.1016/j.synbio.2023.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/30/2023] Open
Abstract
The ability to precisely control activities of engineered designer cells provides a novel strategy for modern precision medicine. Dynamically adjustable gene- and cell-based precision therapies are recognized as next generation medicines. However, the translation of these controllable therapeutics into clinical practice is severely hampered by the lack of safe and highly specific genetic switches controlled by triggers that are nontoxic and side-effect free. Recently, natural products derived from plants have been extensively explored as trigger molecules to control genetic switches and synthetic gene networks for multiple applications. These controlled genetic switches could be further introduced into mammalian cells to obtain synthetic designer cells for adjustable and fine tunable cell-based precision therapy. In this review, we introduce various available natural molecules that were engineered to control genetic switches for controllable transgene expression, complex logic computation, and therapeutic drug delivery to achieve precision therapy. We also discuss current challenges and prospects in translating these natural molecule-controlled genetic switches developed for biomedical applications from the laboratory to the clinic.
Collapse
|
25
|
Li H, Shang Y, Feng Q, Liu Y, Chen J, Dong H. A novel bioartificial pancreas fabricated via islets microencapsulation in anti-adhesive core-shell microgels and macroencapsulation in a hydrogel scaffold prevascularized in vivo. Bioact Mater 2023; 27:362-376. [PMID: 37180642 PMCID: PMC10172916 DOI: 10.1016/j.bioactmat.2023.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Islets transplantation is a promising treatment for type 1 diabetes mellitus. However, severe host immune rejection and poor oxygen/nutrients supply due to the lack of surrounding capillary network often lead to transplantation failure. Herein, a novel bioartificial pancreas is constructed via islets microencapsulation in core-shell microgels and macroencapsulation in a hydrogel scaffold prevascularized in vivo. Specifically, a hydrogel scaffold containing methacrylated gelatin (GelMA), methacrylated heparin (HepMA) and vascular endothelial growth factor (VEGF) is fabricated, which can delivery VEGF in a sustained style and thus induce subcutaneous angiogenesis. In addition, islets-laden core-shell microgels using methacrylated hyaluronic acid (HAMA) as microgel core and poly(ethylene glycol) diacrylate (PEGDA)/carboxybetaine methacrylate (CBMA) as shell layer are prepared, which provide a favorable microenvironment for islets and simultaneously the inhibition of host immune rejection via anti-adhesion of proteins and immunocytes. As a result of the synergistic effect between anti-adhesive core-shell microgels and prevascularized hydrogel scaffold, the bioartificial pancreas can reverse the blood glucose levels of diabetic mice from hyperglycemia to normoglycemia for at least 90 days. We believe this bioartificial pancreas and relevant fabrication method provide a new strategy to treat type 1 diabetes, and also has broad potential applications in other cell therapies.
Collapse
Affiliation(s)
- Haofei Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Yulian Shang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Qi Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Yang Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Junlin Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Hua Dong
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
- Corresponding author. School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
26
|
Beatty R, Mendez KL, Schreiber LHJ, Tarpey R, Whyte W, Fan Y, Robinson ST, O'Dwyer J, Simpkin AJ, Tannian J, Dockery P, Dolan EB, Roche ET, Duffy GP. Soft robot-mediated autonomous adaptation to fibrotic capsule formation for improved drug delivery. Sci Robot 2023; 8:eabq4821. [PMID: 37647382 DOI: 10.1126/scirobotics.abq4821] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
The foreign body response impedes the function and longevity of implantable drug delivery devices. As a dense fibrotic capsule forms, integration of the device with the host tissue becomes compromised, ultimately resulting in device seclusion and treatment failure. We present FibroSensing Dynamic Soft Reservoir (FSDSR), an implantable drug delivery device capable of monitoring fibrotic capsule formation and overcoming its effects via soft robotic actuations. Occlusion of the FSDSR porous membrane was monitored over 7 days in a rodent model using electrochemical impedance spectroscopy. The electrical resistance of the fibrotic capsule correlated to its increase in thickness and volume. Our FibroSensing membrane showed great sensitivity in detecting changes at the abiotic/biotic interface, such as collagen deposition and myofibroblast proliferation. The potential of the FSDSR to overcome fibrotic capsule formation and maintain constant drug dosing over time was demonstrated in silico and in vitro. Controlled closed loop release of methylene blue into agarose gels (with a comparable fold change in permeability relating to 7 and 28 days in vivo) was achieved by adjusting the magnitude and frequency of pneumatic actuations after impedance measurements by the FibroSensing membrane. By sensing fibrotic capsule formation in vivo, the FSDSR will be capable of probing and adapting to the foreign body response through dynamic actuation changes. Informed by real-time sensor signals, this device offers the potential for long-term efficacy and sustained drug dosing, even in the setting of fibrotic capsule formation.
Collapse
Affiliation(s)
- Rachel Beatty
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Keegan L Mendez
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucien H J Schreiber
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Ruth Tarpey
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, School of Engineering, University of Galway, Galway, Ireland
| | - William Whyte
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott T Robinson
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Joanne O'Dwyer
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Joseph Tannian
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Peter Dockery
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Eimear B Dolan
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Biomedical Engineering, School of Engineering, University of Galway, Galway, Ireland
| | - Ellen T Roche
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
- SFI Centre for Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
27
|
Shen M, Zheng L, Koole LH. Polymeric Microspheres Designed to Carry Crystalline Drugs at Their Surface or Inside Cavities and Dimples. Pharmaceutics 2023; 15:2146. [PMID: 37631360 PMCID: PMC10460081 DOI: 10.3390/pharmaceutics15082146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Injectable polymer microparticles with the ability to carry and release pharmacologically active agents are attracting more and more interest. This study is focused on the chemical synthesis, characterization, and preliminary exploration of the utility of a new type of injectable drug-releasing polymer microparticle. The particles feature a new combination of structural and physico-chemical properties: (i) their geometry deviates from the spherical in the sense that the particles have a cavity; (ii) the particles are porous and can therefore be loaded with crystalline drug formulations; drug crystals can reside at both the particle's surfaces and inside cavities; (iii) the particles are relatively dense since the polymer network contains covalently bound iodine (approximately 10% by mass); this renders the drug-loaded particles traceable (localizable) by X-ray fluoroscopy. This study presents several examples. First, the particles were loaded with crystalline voriconazole, which is a potent antifungal drug used in ophthalmology to treat fungal keratitis (infection/inflammation of the cornea caused by penetrating fungus). Drug loading as high as 10% by mass (=mass of immobilized drug/(mass of the microparticle + mass of immobilized drug) × 100%) could be achieved. Slow local release of voriconazole from these particles was observed in vitro. These findings hold promise regarding new approaches to treat fungal keratitis. Moreover, this study can help to expand the scope of the transarterial chemoembolization (TACE) technique since it enables the use of higher drug loadings (thus enabling higher local drug concentration or extended therapy duration), as well as application of hydrophobic drugs that cannot be used in combination with existing TACE embolic particles.
Collapse
Affiliation(s)
| | | | - Leo H. Koole
- Innovative Bioengineering Laboratory for Ocular Drug Delivery, School of Ophthalmology and Optometry, Eye Hospital of Wenzhou Medical University, Wenzhou Medical University, 270 Xueyuan West Road, Wenzhou 325027, China; (M.S.); (L.Z.)
| |
Collapse
|
28
|
Ren Y, Chen Y, Chen W, Deng H, Li P, Liu Y, Gao C, Tian G, Ning C, Yuan Z, Sui X, Liu S, Guo Q. Hydrophilic nanofibers with aligned topography modulate macrophage-mediated host responses via the NLRP3 inflammasome. J Nanobiotechnology 2023; 21:269. [PMID: 37574546 PMCID: PMC10424429 DOI: 10.1186/s12951-023-02024-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/24/2023] [Indexed: 08/15/2023] Open
Abstract
Successful biomaterial implantation requires appropriate immune responses. Macrophages are key mediators involved in this process. Currently, exploitation of the intrinsic properties of biomaterials to modulate macrophages and immune responses is appealing. In this study, we prepared hydrophilic nanofibers with an aligned topography by incorporating polyethylene glycol and polycaprolactone using axial electrospinning. We investigated the effect of the nanofibers on macrophage behavior and the underlying mechanisms. With the increase of hydrophilicity of aligned nanofibers, the inflammatory gene expression of macrophages adhering to them was downregulated, and M2 polarization was induced. We further presented clear evidence that the inflammasome NOD-like receptor thermal protein domain associated protein 3 (NLRP3) was the cellular sensor by which macrophages sense the biomaterials, and it acted as a regulator of the macrophage-mediated response to foreign bodies and implant integration. In vivo, we showed that the fibers shaped the implant-related immune microenvironment and ameliorated peritendinous adhesions. In conclusion, our study demonstrated that hydrophilic aligned nanofibers exhibited better biocompatibility and immunological properties.
Collapse
Affiliation(s)
- Yiming Ren
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yi Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Wei Chen
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Haotian Deng
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Peiqi Li
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yubo Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Cangjian Gao
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Guangzhao Tian
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Chao Ning
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Zhiguo Yuan
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiang Sui
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shuyun Liu
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Institute of Orthopedics, First Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
29
|
Tawagi E, Ung T, Cheng HLM, Santerre JP. Arrhenius-model-based degradable oligourethane hydrogels for controlled growth factor release. Acta Biomater 2023; 166:167-186. [PMID: 37207744 DOI: 10.1016/j.actbio.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/22/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
Biodegradable hydrogels are growing in demand to enable the delivery of biomolecules (e.g. growth factors) for regenerative medicine. This research investigated the resorption of an oligourethane/polyacrylic acid hydrogel, a biodegradable hydrogel which supports tissue regeneration. The Arrhenius model was used to characterize the resorption of the polymeric gels in relevant in vitro conditions, and the Flory-Rehner equation was used to correlate the volumetric swelling ratio with the extent of degradation. The study found that the swelling rate of the hydrogel follows the Arrhenius model at elevated temperatures, estimating degradation time in saline solution at 37°C to be between 5 and 13 months, serving as a preliminary approximation of degradation in vivo. The degradation products had low cytotoxicity towards endothelial cells, and the hydrogel supported stromal cell proliferation. Additionally, the hydrogels were able to release growth factors and maintain the biomolecules' bioactivity towards cell proliferation. The study of the vascular endothelial growth factor (VEGF) release from the hydrogel used a diffusion process model, showing that the electrostatic attraction between VEGF and the anionic hydrogel allowed for controlled and sustained VEGF release over three weeks. In a rat subcutaneous implant model, a selected hydrogel with desired degradation rates exhibited minimal foreign body response and supported M2a macrophage phenotype, and vascularization. The low M1 and high M2a macrophage phenotypes within the implants were associated with tissue integration. This research supports the use of oligourethane/polyacrylic acid hydrogels as a promising material for delivering growth factors and supporting tissue regeneration. STATEMENT OF SIGNIFICANCE: There is a need for degradable elastomeric hydrogels that can support the formation of soft tissues and minimize long-term foreign body responses. An Arrhenius model was used to estimate the relative breakdown of hydrogels, in-vitro. The results demonstrate that hydrogels made from a combination of poly(acrylic acid) and oligo-urethane diacrylates can be designed to resorb over defined periods ranging from months to years depending on the chemical formulation prescribed by the model. The hydrogel formulations also provided for different release profiles of growth factors, relevant to tissue regeneration. In-vivo, these hydrogels had minimal inflammatory effects and showed evidence of integration into the surrounding tissue. The hydrogel approach can help the field design a broader range of biomaterials for tissue regeneration.
Collapse
Affiliation(s)
- Eric Tawagi
- Institute of Biomedical Engineering, University of Toronto, 661 University Avenue, 14th Floor, Room 1435, Toronto, ON M5G 1M1, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Trevor Ung
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Hai-Ling Margaret Cheng
- Institute of Biomedical Engineering, University of Toronto, 661 University Avenue, 14th Floor, Room 1435, Toronto, ON M5G 1M1, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada; The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, ON, Canada
| | - J Paul Santerre
- Institute of Biomedical Engineering, University of Toronto, 661 University Avenue, 14th Floor, Room 1435, Toronto, ON M5G 1M1, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Jeon S, Lee YS, Oh SR, Jeong J, Lee DH, So KH, Hwang NS. Recent advances in endocrine organoids for therapeutic application. Adv Drug Deliv Rev 2023; 199:114959. [PMID: 37301512 DOI: 10.1016/j.addr.2023.114959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/21/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
The endocrine system, consisting of the hypothalamus, pituitary, endocrine glands, and hormones, plays a critical role in hormone metabolic interactions. The complexity of the endocrine system is a significant obstacle to understanding and treating endocrine disorders. Notably, advances in endocrine organoid generation allow a deeper understanding of the endocrine system by providing better comprehension of molecular mechanisms of pathogenesis. Here, we highlight recent advances in endocrine organoids for a wide range of therapeutic applications, from cell transplantation therapy to drug toxicity screening, combined with development in stem cell differentiation and gene editing technologies. In particular, we provide insights into the transplantation of endocrine organoids to reverse endocrine dysfunctions and progress in developing strategies for better engraftments. We also discuss the gap between preclinical and clinical research. Finally, we provide future perspectives for research on endocrine organoids for the development of more effective treatments for endocrine disorders.
Collapse
Affiliation(s)
- Suwan Jeon
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sun Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seh Ri Oh
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinseong Jeong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Hyun Lee
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung-Ha So
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea.
| | - Nathaniel S Hwang
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea; Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
31
|
Desai P, Dasgupta A, Sofias AM, Peña Q, Göstl R, Slabu I, Schwaneberg U, Stiehl T, Wagner W, Jockenhövel S, Stingl J, Kramann R, Trautwein C, Brümmendorf TH, Kiessling F, Herrmann A, Lammers T. Transformative Materials for Interfacial Drug Delivery. Adv Healthc Mater 2023; 12:e2301062. [PMID: 37282805 PMCID: PMC11468550 DOI: 10.1002/adhm.202301062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/25/2023] [Indexed: 06/08/2023]
Abstract
Drug delivery systems (DDS) are designed to temporally and spatially control drug availability and activity. They assist in improving the balance between on-target therapeutic efficacy and off-target toxic side effects. DDS aid in overcoming biological barriers encountered by drug molecules upon applying them via various routes of administration. They are furthermore increasingly explored for modulating the interface between implanted (bio)medical materials and host tissue. Herein, an overview of the biological barriers and host-material interfaces encountered by DDS upon oral, intravenous, and local administration is provided, and material engineering advances at different time and space scales to exemplify how current and future DDS can contribute to improved disease treatment are highlighted.
Collapse
Affiliation(s)
- Prachi Desai
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
| | - Quim Peña
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Robert Göstl
- DWI – Leibniz Institute for Interactive Materials52074AachenGermany
| | - Ioana Slabu
- Institute of Applied Medical EngineeringHelmholtz InstituteMedical FacultyRWTH Aachen University52074AachenGermany
| | | | - Thomas Stiehl
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Institute for Computational Biomedicine – Disease ModelingRWTH Aachen University52074AachenGermany
| | - Wolfgang Wagner
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
- Institute for Stem Cell BiologyUniversity Hospital of RWTH Aachen52074AachenGermany
| | - Stefan Jockenhövel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz Institute AachenRWTH Aachen University52074AachenGermany
| | - Julia Stingl
- Institute of Clinical PharmacologyUniversity Hospital RWTH Aachen52074AachenGermany
| | - Rafael Kramann
- Division of Nephrology and Clinical ImmunologyRWTH Aachen University52074AachenGermany
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University52074AachenGermany
| | - Christian Trautwein
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Department of Medicine III (GastroenterologyMetabolic diseases and Intensive Care)University Hospital RWTH Aachen52074AachenGermany
| | - Tim H. Brümmendorf
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Department of HematologyOncologyHemostaseology and Stem Cell TransplantationRWTH Aachen University Medical School52074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
| | - Andreas Herrmann
- DWI – Leibniz Institute for Interactive Materials52074AachenGermany
- Institute of Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 152074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
| |
Collapse
|
32
|
Lu K, Brauns T, Sluder AE, Poznansky MC, Dogan F. Combinatorial islet protective therapeutic approaches in β-cell transplantation: Rationally designed solutions using a target product profile. FASEB Bioadv 2023; 5:287-304. [PMID: 37415930 PMCID: PMC10320848 DOI: 10.1096/fba.2023-00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 07/08/2023] Open
Abstract
While progress has been made in the development of islet cell transplantation (ICT) as a viable alternative to the use of exogenous insulin therapy in the treatment of type 1 diabetes, it has not yet achieved its full potential in clinical studies. Ideally, ICT would enable lifelong maintenance of euglycemia without the need for exogenous insulin, blood glucose monitoring or systemic immune suppression. To achieve such an optimal result, therapeutic approaches should simultaneously promote long-term islet viability, functionality, and localized immune protection. In practice, however, these factors are typically tackled individually. Furthermore, while the requirements of optimal ICT are implicitly acknowledged across numerous publications, the literature contains few comprehensive articulations of the target product profile (TPP) for an optimal ICT product, including key characteristics of safety and efficacy. This review aims to provide a novel TPP for ICT and presents promising tried and untried combinatorial approaches that could be used to achieve the target product profile. We also highlight regulatory barriers to the development and adoption of ICT, particularly in the United States, where ICT is only approved for use in academic clinical trials and is not reimbursed by insurance carriers. Overall, this review argues that the clear definition of a TPP in addition to the use of combinatorial approaches could help to overcome the clinical barriers to the widespread adoption of ICT for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Katie Lu
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
- Department of BiologyStanford UniversityStanfordCaliforniaUSA
| | - Timothy Brauns
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| | - Ann E. Sluder
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| | - Fatma Dogan
- Vaccine and Immunotherapy CenterMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
33
|
Mukherjee S, Kim B, Cheng LY, Doerfert MD, Li J, Hernandez A, Liang L, Jarvis MI, Rios PD, Ghani S, Joshi I, Isa D, Ray T, Terlier T, Fell C, Song P, Miranda RN, Oberholzer J, Zhang DY, Veiseh O. Screening hydrogels for antifibrotic properties by implanting cellularly barcoded alginates in mice and a non-human primate. Nat Biomed Eng 2023; 7:867-886. [PMID: 37106151 PMCID: PMC10593184 DOI: 10.1038/s41551-023-01016-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 02/27/2023] [Indexed: 04/29/2023]
Abstract
Screening implantable biomaterials for antifibrotic properties is constrained by the need for in vivo testing. Here we show that the throughput of in vivo screening can be increased by cellularly barcoding a chemically modified combinatorial library of hydrogel formulations. The method involves the implantation of a mixture of alginate formulations, each barcoded with human umbilical vein endothelial cells from different donors, and the association of the identity and performance of each formulation by genotyping single nucleotide polymorphisms of the cells via next-generation sequencing. We used the method to screen 20 alginate formulations in a single mouse and 100 alginate formulations in a single non-human primate, and identified three lead hydrogel formulations with antifibrotic properties. Encapsulating human islets with one of the formulations led to long-term glycaemic control in a mouse model of diabetes, and coating medical-grade catheters with the other two formulations prevented fibrotic overgrowth. High-throughput screening of barcoded biomaterials in vivo may help identify formulations that enhance the long-term performance of medical devices and of biomaterial-encapsulated therapeutic cells.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX, USA
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Lauren Y Cheng
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Jiaming Li
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Lily Liang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | | | | | - Trisha Ray
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Tanguy Terlier
- SIMS Laboratory, Shared Equipment Authority, Rice University, Houston, TX, USA
| | - Cody Fell
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Ping Song
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Roberto N Miranda
- Department of Hematopathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jose Oberholzer
- Division of Transplant Surgery, University of Virginia, Charlottesville, VA, USA
| | - David Yu Zhang
- Department of Bioengineering, Rice University, Houston, TX, USA.
- NuProbe USA, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
34
|
Qin T, Smink AM, de Vos P. Enhancing longevity of immunoisolated pancreatic islet grafts by modifying both the intracapsular and extracapsular environment. Acta Biomater 2023:S1742-7061(23)00362-8. [PMID: 37392934 DOI: 10.1016/j.actbio.2023.06.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic metabolic disease characterized by autoimmune destruction of pancreatic β cells. Transplantation of immunoisolated pancreatic islets might treat T1DM in the absence of chronic immunosuppression. Important advances have been made in the past decade as capsules can be produced that provoke minimal to no foreign body response after implantation. However, graft survival is still limited as islet dysfunction may occur due to chronic damage to islets during islet isolation, immune responses induced by inflammatory cells, and nutritional issues for encapsulated cells. This review summarizes the current challenges for promoting longevity of grafts. Possible strategies for improving islet graft longevity are also discussed, including supplementation of the intracapsular milieu with essential survival factors, promotion of vascularization and oxygenation near capsules, modulation of biomaterials, and co-transplantation of accessory cells. Current insight is that both the intracapsular as well as the extracapsular properties should be improved to achieve long-term survival of islet-tissue. Some of these approaches reproducibly induce normoglycemia for more than a year in rodents. Further development of the technology requires collective research efforts in material science, immunology, and endocrinology. STATEMENT OF SIGNIFICANCE: Islet immunoisolation allows for transplantation of insulin producing cells in absence of immunosuppression and might facilitate the use of xenogeneic cell sources or grafting of cells obtained from replenishable cell sources. However, a major challenge to date is to create a microenvironment that supports long-term graft survival. This review provides a comprehensive overview of the currently identified factors that have been demonstrated to be involved in either stimulating or reducing islet graft survival in immunoisolating devices and discussed current strategies to enhance the longevity of encapsulated islet grafts as treatment for type 1 diabetes. Although significant challenges remain, interdisciplinary collaboration across fields may overcome obstacles and facilitate the translation of encapsulated cell therapy from the laboratory to clinical application.
Collapse
Affiliation(s)
- Tian Qin
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands.
| | - Alexandra M Smink
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
35
|
Doloff JC, Ma M, Sadraei A, Tam HH, Farah S, Hollister-Lock J, Vegas AJ, Veiseh O, Quiroz VM, Rakoski A, Aresta-DaSilva S, Bader AR, Griffin M, Weir GC, Brehm MA, Shultz LD, Langer R, Greiner DL, Anderson DG. Identification of a humanized mouse model for functional testing of immune-mediated biomaterial foreign body response. SCIENCE ADVANCES 2023; 9:eade9488. [PMID: 37327334 PMCID: PMC10275594 DOI: 10.1126/sciadv.ade9488] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 05/05/2023] [Indexed: 06/18/2023]
Abstract
Biomedical devices comprise a major component of modern medicine, however immune-mediated fibrosis and rejection can limit their function over time. Here, we describe a humanized mouse model that recapitulates fibrosis following biomaterial implantation. Cellular and cytokine responses to multiple biomaterials were evaluated across different implant sites. Human innate immune macrophages were verified as essential to biomaterial rejection in this model and were capable of cross-talk with mouse fibroblasts for collagen matrix deposition. Cytokine and cytokine receptor array analysis confirmed core signaling in the fibrotic cascade. Foreign body giant cell formation, often unobserved in mice, was also prominent. Last, high-resolution microscopy coupled with multiplexed antibody capture digital profiling analysis supplied spatial resolution of rejection responses. This model enables the study of human immune cell-mediated fibrosis and interactions with implanted biomaterials and devices.
Collapse
Affiliation(s)
- Joshua C. Doloff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Minglin Ma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Atieh Sadraei
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Hok Hei Tam
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Shady Farah
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Jennifer Hollister-Lock
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | - Arturo J. Vegas
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Omid Veiseh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Victor M. Quiroz
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Amanda Rakoski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Stephanie Aresta-DaSilva
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Andrew R. Bader
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
| | - Marissa Griffin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
| | - Gordon C. Weir
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | - Michael A. Brehm
- Program in Molecular Medicine, Diabetes Centre of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Dale L. Greiner
- Program in Molecular Medicine, Diabetes Centre of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St., Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| |
Collapse
|
36
|
Hogrebe NJ, Ishahak M, Millman JR. Developments in stem cell-derived islet replacement therapy for treating type 1 diabetes. Cell Stem Cell 2023; 30:530-548. [PMID: 37146579 PMCID: PMC10167558 DOI: 10.1016/j.stem.2023.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
The generation of islet-like endocrine clusters from human pluripotent stem cells (hPSCs) has the potential to provide an unlimited source of insulin-producing β cells for the treatment of diabetes. In order for this cell therapy to become widely adopted, highly functional and well-characterized stem cell-derived islets (SC-islets) need to be manufactured at scale. Furthermore, successful SC-islet replacement strategies should prevent significant cell loss immediately following transplantation and avoid long-term immune rejection. This review highlights the most recent advances in the generation and characterization of highly functional SC-islets as well as strategies to ensure graft viability and safety after transplantation.
Collapse
Affiliation(s)
- Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA.
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA.
| |
Collapse
|
37
|
Li L, Shapiro RL, Joo MK, Josyula A, Hsueh HT, Gutierrez OB, Halpert G, Akshintala V, Chen H, Curtis S, Better M, Davison C, Hu H, Almario JAN, Steinway SN, Hunt K, Del Sesto RE, Izzi J, Salimian KJ, Ensign LM, Selaru FM. Injectable, Drug-Eluting Nanocrystals Prevent Fibrosis and Stricture Formation In Vivo. Gastroenterology 2023; 164:937-952.e13. [PMID: 36657529 PMCID: PMC10151160 DOI: 10.1053/j.gastro.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/07/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS Tissue fibrosis results from uncontrolled healing responses leading to excessive mesenchymal cell activation and collagen and other extracellular matrix deposition. In the gastrointestinal tract, fibrosis leads to narrowing of the lumen and stricture formation. A drug treatment to prevent fibrosis and strictures in the gastrointestinal tract would be transformational for patient care. We aimed to develop a stricture treatment with the following characteristics and components: a small molecule with strong antifibrotic effects that is delivered locally at the site of the stricture to ensure correct lesional targeting while protecting the systemic circulation, and that is formulated with sustained-release properties to act throughout the wound healing processes. METHODS A high-throughput drug screening was performed to identify small molecules with antifibrotic properties. Next, we formulated an antifibrotic small molecule for sustained release and tested its antifibrotic potential in 3 animal models of fibrosis. RESULTS Sulconazole, a US Food and Drug Administration-approved drug for fungal infections, was found to have strong antifibrotic properties. Sulconazole was formulated as sulconazole nanocrystals for sustained release. We found that sulconazole nanocrystals provided superior or equivalent fibrosis prevention with less frequent dosing in mouse models of skin and intestinal tissue fibrosis. In a patient-like swine model of bowel stricture, a single injection of sulconazole nanocrystals prevented stricture formation. CONCLUSIONS The current data lay the foundation for further studies to improve the management of a range of diseases and conditions characterized by tissue fibrosis.
Collapse
Affiliation(s)
- Ling Li
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Rachel L Shapiro
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Min Kyung Joo
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aditya Josyula
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Henry T Hsueh
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Olaya Brewer Gutierrez
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Gilad Halpert
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Venkata Akshintala
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Haiming Chen
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Samuel Curtis
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marina Better
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charlotte Davison
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Haijie Hu
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Jose Antonio Navarro Almario
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Steven N Steinway
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Kelton Hunt
- Department of Chemistry and Biochemistry, Utah Tech University, St George, Utah
| | - Rico E Del Sesto
- Department of Chemistry and Biochemistry, Utah Tech University, St George, Utah
| | - Jessica Izzi
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland
| | - Kevan J Salimian
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura M Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Gynecology and Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Medicine, Division of Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Florin M Selaru
- Division of Gastroenterology and Hepatology, School of Medicine, The Johns Hopkins University, Baltimore, Maryland; Department of Oncology, Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland; The Institute for Nanobiotechnology, The Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
38
|
Santanelli di Pompeo F, Sorotos M, Canese R, Valeri M, Roberto C, Giorgia S, Firmani G, di Napoli A. Study of the Effect of Different Breast Implant Surfaces on Capsule Formation and Host Inflammatory Response in an Animal Model. Aesthet Surg J 2023; 43:506-515. [PMID: 36402143 DOI: 10.1093/asj/sjac301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Breast implants are biomaterials eliciting a physiological and mandatory foreign body response. OBJECTIVES The authors designed an animal study to investigate the impact of different implant surfaces on the formation of the periprosthetic capsule, the inflammatory response, and the cellular composition. METHODS The authors implanted 1 scaled-down version of breast implants by different manufactures on 70 female Sprague Dawley rats. Animals were divided into 5 groups of 14 animals. Group A received a smooth implant (Ra ≈ 0.5 µm) according to the ISO 14607-2018 classification, Group B a smooth implant (Ra ≈ 3.2 µm), Group C a smooth implant (Ra ≈ 5 µm), Group D a macrotextured implant (Ra ≈ 62 µm), and Group E a macrotextured implant (Ra ≈ 75 µm). At 60 days, all animals received a magnetic resonance imaging (MRI), and 35 animals were killed and their capsules sent for histology (capsule thickness, inflammatory infiltrate) and immunohistochemistry analysis (cellular characterization). The remaining animals repeated the MRI at 120 days and were killed following the same protocol. RESULTS MRI showed a thinner capsule in the smooth implants (Groups A-C) at 60 days (P < .001) but not at 120 days (P = .039), confirmed with histology both at 60 days (P = .005) and 120 days (P < .001). Smooth implants (Groups A-C) presented a mild inflammatory response at 60 days that was maintained at 120 days and a high M2-Macrophage concentration (anti-inflammatory). CONCLUSIONS Our study confirms that smooth implants form a thinner capsule, inferior inflammatory infiltrate, and a cellular composition that indicates a mild host inflammatory response. A new host inflammatory response classification is elaborated classifying breast implants into mild, moderate, and high.
Collapse
|
39
|
Bian N, Chu C, Rung S, Huangphattarakul V, Man Y, Lin J, Hu C. Immunomodulatory Biomaterials and Emerging Analytical Techniques for Probing the Immune Micro-Environment. Tissue Eng Regen Med 2023; 20:11-24. [PMID: 36241939 PMCID: PMC9852373 DOI: 10.1007/s13770-022-00491-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 02/01/2023] Open
Abstract
After implantation of a biomaterial, both the host immune system and properties of the material determine the local immune response. Through triggering or modulating the local immune response, materials can be designed towards a desired direction of promoting tissue repair or regeneration. High-throughput sequencing technologies such as single-cell RNA sequencing (scRNA-seq) emerging as a powerful tool for dissecting the immune micro-environment around biomaterials, have not been fully utilized in the field of soft tissue regeneration. In this review, we first discussed the procedures of foreign body reaction in brief. Then, we summarized the influences that physical and chemical modulation of biomaterials have on cell behaviors in the micro-environment. Finally, we discussed the application of scRNA-seq in probing the scaffold immune micro-environment and provided some reference to designing immunomodulatory biomaterials. The foreign body response consists of a series of biological reactions. Immunomodulatory materials regulate immune cell activation and polarization, mediate divergent local immune micro-environments and possess different tissue engineering functions. The manipulation of physical and chemical properties of scaffolds can modulate local immune responses, resulting in different outcomes of fibrosis or tissue regeneration. With the advancement of technology, emerging techniques such as scRNA-seq provide an unprecedented understanding of immune cell heterogeneity and plasticity in a scaffold-induced immune micro-environment at high resolution. The in-depth understanding of the interaction between scaffolds and the host immune system helps to provide clues for the design of biomaterials to optimize regeneration and promote a pro-regenerative local immune micro-environment.
Collapse
Affiliation(s)
- Nanyan Bian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenyu Chu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Shengan Rung
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Vicha Huangphattarakul
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Jie Lin
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| | - Chen Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
40
|
Huang P, Xu J, Xie L, Gao G, Chen S, Gong Z, Lao X, Shan Z, Shi J, Zhou Z, Chen Z, Cao Y, Wang Y, Chen Z. Improving hard metal implant and soft tissue integration by modulating the “inflammatory-fibrous complex” response. Bioact Mater 2023; 20:42-52. [PMID: 35633873 PMCID: PMC9127122 DOI: 10.1016/j.bioactmat.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/23/2022] [Accepted: 05/08/2022] [Indexed: 11/29/2022] Open
|
41
|
Audouard E, Michel F, Pierroz V, Kim T, Rousselot L, Gillet-Legrand B, Dufayet-Chauffaut G, Buchmann P, Florea M, Khel A, Altynbekova K, Delgaldo C, Escudero E, Soler ABA, Cartier N, Piguet F, Folcher M. Bioelectronic cell-based device provides a strategy for the treatment of the experimental model of multiple sclerosis. J Control Release 2022; 352:994-1008. [PMID: 36370877 PMCID: PMC9733677 DOI: 10.1016/j.jconrel.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022]
Abstract
Wireless powered optogenetic cell-based implant provides a strategy to deliver subcutaneously therapeutic proteins. Immortalize Human Mesenchymal Stem Cells (hMSC-TERT) expressing the bacteriophytochrome diguanylate cyclase (DGCL) were validated for optogenetic controlled interferon-β delivery (Optoferon cells) in a bioelectronic cell-based implant. Optoferon cells transcriptomic profiling was used to elaborate an in-silico model of the recombinant interferon-β production. Wireless optoelectronic device integration was developed using additive manufacturing and injection molding. Implant cell-based optoelectronic interface manufacturing was established to integrate industrial flexible compact low-resistance screen-printed Near Field Communication (NFC) coil antenna. Optogenetic cell-based implant biocompatibility, and device performances were evaluated in the Experimental Autoimmune Encephalomyelitis (EAE) mouse model of multiple sclerosis.
Collapse
Affiliation(s)
- Emilie Audouard
- NeuroGenCell, Paris Brain Institute – ICM, INSERM, CNRS, AP-HP, Sorbonne Université; Hôpital de la Pitié Salpêtrière, Paris, France
| | - Fanny Michel
- Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland
| | - Vanessa Pierroz
- Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland
| | - Taeuk Kim
- Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland
| | - Lisa Rousselot
- NeuroGenCell, Paris Brain Institute – ICM, INSERM, CNRS, AP-HP, Sorbonne Université; Hôpital de la Pitié Salpêtrière, Paris, France
| | - Béatrix Gillet-Legrand
- NeuroGenCell, Paris Brain Institute – ICM, INSERM, CNRS, AP-HP, Sorbonne Université; Hôpital de la Pitié Salpêtrière, Paris, France
| | - Gaëlle Dufayet-Chauffaut
- NeuroGenCell, Paris Brain Institute – ICM, INSERM, CNRS, AP-HP, Sorbonne Université; Hôpital de la Pitié Salpêtrière, Paris, France
| | - Peter Buchmann
- Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland
| | - Michael Florea
- Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland
| | | | | | - Claudia Delgaldo
- Eurecat, Centre Tecnològic de Catalunya, Functional Printing and Embedded Devices Unit, Mataró, Spain
| | - Encarna Escudero
- Eurecat, Centre Tecnològic de Catalunya, Functional Printing and Embedded Devices Unit, Mataró, Spain
| | - Alejandra Ben Aissa Soler
- Eurecat, Centre Tecnològic de Catalunya, Functional Printing and Embedded Devices Unit, Mataró, Spain
| | - Nathalie Cartier
- NeuroGenCell, Paris Brain Institute – ICM, INSERM, CNRS, AP-HP, Sorbonne Université; Hôpital de la Pitié Salpêtrière, Paris, France
| | - Francoise Piguet
- NeuroGenCell, Paris Brain Institute – ICM, INSERM, CNRS, AP-HP, Sorbonne Université; Hôpital de la Pitié Salpêtrière, Paris, France
| | - Marc Folcher
- Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland,Institute of Molecular and Clinical Ophthalmology, IOB, Basel, Switzerland,Corresponding author at: Department of Biosystems Science and Engineering, D-BSSE, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
42
|
Different Molecular Features of Epithelioid and Giant Cells in Foreign Body Reaction Identified by Single-Cell RNA Sequencing. J Invest Dermatol 2022; 142:3232-3242.e16. [PMID: 35853485 DOI: 10.1016/j.jid.2022.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 01/05/2023]
Abstract
Although macrophage‒epithelioid cell (EPC)‒giant cell (GC) differentiation is acknowledged in foreign body reaction (FBR), the exact molecular features remain elusive. To discover the molecular profiles of EPC and GC, we analyzed mouse sponge and silk FBRs by integrating single-cell RNA sequencing and spatial sequencing, which identified seven cell types, including macrophages and fibroblasts. Macrophages comprised three subsets with a trajectory from M2-like cell to EPC to GC. They were different in many aspects, including cytokine, extracellular matrix organization/degradation, epithelial modules, and glycolysis that were consistent in both sponge and silk FBRs. EPCs exhibited epithelial modules and extracellular matrix organization, and GCs showed glycolysis, extracellular matrix degradation, and cell fusion signatures. Cellular interactions in GCs and M2-like cells were predicted to be higher than that in EPCs. High expression of inflammation or fusion-related (GPNMB, matrix metalloproteinase 12 gene MMP12, DCSTAMP) and glycolysis-related (PGAM1, ALDOA) genes was identified in GCs of human/mouse tissues, suggesting them as GC-specific markers. Our study identified unique signatures of EPCs and GCs in FBR. Importantly, GCs showed strong glycolysis signatures and cellular interactions, suggesting their activation in FBR. Our data on EPC and GC refinement and GC-specific markers enable the understanding of FBR and help to explore preventive and therapeutic management strategies for skin FBRs.
Collapse
|
43
|
Surface configuration of microarc oxidized Ti with regionally loaded chitosan hydrogel containing ciprofloxacin for improving biological performance. Mater Today Bio 2022; 16:100380. [PMID: 36033377 PMCID: PMC9399291 DOI: 10.1016/j.mtbio.2022.100380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/12/2022] [Accepted: 07/21/2022] [Indexed: 11/23/2022] Open
Abstract
The bacterial colonization and poor osseointegration of Ti implants significantly compromise their applications in load-bearing bone repair and replacement. To endorse the Ti with both excellent bioactivity and antibacterial ability, we developed a microarc oxidation coating that was modified uniformly by hydroxyapatite (HA) nanodots arrays and loaded regionally with chitosan hydrogel containing ciprofloxacin. The bonding between the HA nanodots covered coating and the chitosan hydrogel is further enhanced via silanization and chemical grafting of glutaraldehyde. Benefiting from the regionally loaded structure of the chitosan hydrogel, the chitosan hydrogel unloaded area can promote the cell adhesion and proliferation with excellent bioactivity, though relatively low OD value of cck8 has been observed at the beginning of the cell culturing. Whereas, the OD value of cck8 rises with the prolongation of the cell culturing time due to the degradation of the regionally loaded chitosan hydrogel. With the help of the laden ciprofloxacin in chitosan hydrogels, the sample effectively sterilizes the bacterial with a bacteriostatic ring. Therefore, regional loading of chitosan hydrogel containing ciprofloxacin on the modified microarc oxidation coating is a good approach to endorse Ti with both excellent bioactivity and antibacterial ability.
Collapse
|
44
|
Moise S, Dolcetti L, Dazzi F, Roach P, Buttery L, MacNeil S, Medcalf N. Assessing the immunosuppressive activity of alginate-encapsulated mesenchymal stromal cells on splenocytes. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:168-176. [PMID: 35726746 DOI: 10.1080/21691401.2022.2088547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/09/2022] [Accepted: 06/05/2022] [Indexed: 06/15/2023]
Abstract
Mesenchymal stromal cells (MSCs) show immunosuppressive effects both via cell-to-cell contact (direct) with immune cells and by producing paracrine factors and extracellular vesicles (indirect). A key challenge in delivering this therapeutic effect in vivo is retaining the MSCs at the site of injection. One way to address this is by encapsulating the MSCs within suitable biomaterial scaffolds. Here, we assess the immunosuppressive effect of alginate-encapsulated murine MSCs on proliferating murine splenocytes. Our results show that MSCs are able to significantly suppress splenocyte proliferation by ∼50% via the indirect mechanism and almost completely (∼98%) via the direct mechanism. We also show for the first time that MSCs as monolayers on tissue culture plastic or encapsulated within alginate, when physically isolated from the splenocytes via transwells, are able to sustain immunosuppressive activity with repeated exposure to fresh splenocytes, for as long as 9 days. These results indicate the need to identify design strategies to simultaneously deliver both modes of MSC immunosuppression. By designing cell-biomaterial constructs with tailored degradation profiles, we can achieve a more sustained (avoiding MSCs migration and apoptosis) and controlled release of both the paracrine signals and eventually the cells themselves enabling efficient MSC-based immunosuppressive therapies for wound healing.
Collapse
Affiliation(s)
- Sandhya Moise
- Centre for Integrated Bioprocessing Research (CIBR), Department of Chemical Engineering, University of Bath, Bath, UK
- Centre for Therapeutic Innovation (CTI), University of Bath, Bath, UK
| | - Luigi Dolcetti
- Department of Medicine and Pharmaceutical Science, King's College London, London, UK
| | - Francesco Dazzi
- Department of Haematological malignancies and stem cell transplant, Kings College hospital NHS trust, London, UK
| | - Paul Roach
- Department of Chemistry, Loughborough University, Loughborough, UK
| | - Lee Buttery
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Sheila MacNeil
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, UK
| | - Nick Medcalf
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough, UK
| |
Collapse
|
45
|
Wu J, Shaidani S, Theodossiou SK, Hartzell EJ, Kaplan DL. Localized, on-demand, sustained drug delivery from biopolymer-based materials. Expert Opin Drug Deliv 2022; 19:1317-1335. [PMID: 35930000 PMCID: PMC9617770 DOI: 10.1080/17425247.2022.2110582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Local drug delivery facilitiates higher concentrations of drug molecules at or near the treatment site to enhance treatment efficiency and reduce drug toxicity and other systemic side effects. However, local drug delivery systems face challenges in terms of encapsulation, delivery, and controlled release of therapeutics. AREAS COVERED We provide an overview of naturally derived biopolymer-based drug delivery systems for localized, sustained, and on-demand treatment. We introduce the advantages and limitations of these systems for drug encapsulation, delivery, and local release, as well as recent applications. EXPERT OPINION Naturally derived biopolymers like cellulose, silk fibroin, chitosan, alginate, hyaluronic acid, and gelatin are good candidates for localized drug delivery because they are readily chemically modified, biocompatible, biodegradable (with the generation of metabolically compatible degradation products), and can be processed in aqueous and ambient environments to maintain the bioactivity of various therapeutics. The tradeoff between the effective treatment dosage and the response by local healthy tissue should be balanced during the design of these delivery systems. Future directions will be focused on strategies to design tunable and controlled biodegradation rates, as well as to explore commercial utility in substituting biopolymer-based systems for currently utilized synthetic polymers for implants for drug delivery.
Collapse
Affiliation(s)
- Junqi Wu
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, USA, 02155
| | - Sawnaz Shaidani
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, USA, 02155
| | - Sophia K. Theodossiou
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, USA, 02155
| | - Emily J. Hartzell
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, USA, 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, USA, 02155
| |
Collapse
|
46
|
Bashor CJ, Hilton IB, Bandukwala H, Smith DM, Veiseh O. Engineering the next generation of cell-based therapeutics. Nat Rev Drug Discov 2022; 21:655-675. [PMID: 35637318 PMCID: PMC9149674 DOI: 10.1038/s41573-022-00476-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 12/19/2022]
Abstract
Cell-based therapeutics are an emerging modality with the potential to treat many currently intractable diseases through uniquely powerful modes of action. Despite notable recent clinical and commercial successes, cell-based therapies continue to face numerous challenges that limit their widespread translation and commercialization, including identification of the appropriate cell source, generation of a sufficiently viable, potent and safe product that meets patient- and disease-specific needs, and the development of scalable manufacturing processes. These hurdles are being addressed through the use of cutting-edge basic research driven by next-generation engineering approaches, including genome and epigenome editing, synthetic biology and the use of biomaterials.
Collapse
Affiliation(s)
- Caleb J Bashor
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Biosciences, Rice University, Houston, TX, USA.
| | - Isaac B Hilton
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Biosciences, Rice University, Houston, TX, USA.
| | - Hozefa Bandukwala
- Sigilon Therapeutics, Cambridge, MA, USA
- Flagship Pioneering, Cambridge, MA, USA
| | - Devyn M Smith
- Sigilon Therapeutics, Cambridge, MA, USA
- Arbor Biotechnologies, Cambridge, MA, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
47
|
Capuani S, Malgir G, Chua CYX, Grattoni A. Advanced strategies to thwart foreign body response to implantable devices. Bioeng Transl Med 2022; 7:e10300. [PMID: 36176611 PMCID: PMC9472022 DOI: 10.1002/btm2.10300] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Abstract
Mitigating the foreign body response (FBR) to implantable medical devices (IMDs) is critical for successful long-term clinical deployment. The FBR is an inevitable immunological reaction to IMDs, resulting in inflammation and subsequent fibrotic encapsulation. Excessive fibrosis may impair IMDs function, eventually necessitating retrieval or replacement for continued therapy. Therefore, understanding the implant design parameters and their degree of influence on FBR is pivotal to effective and long lasting IMDs. This review gives an overview of FBR as well as anti-FBR strategies. Furthermore, we highlight recent advances in biomimetic approaches to resist FBR, focusing on their characteristics and potential biomedical applications.
Collapse
Affiliation(s)
- Simone Capuani
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- University of Chinese Academy of Science (UCAS)BeijingChina
| | - Gulsah Malgir
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- Department of Biomedical EngineeringUniversity of HoustonHoustonTexasUSA
| | | | - Alessandro Grattoni
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- Department of SurgeryHouston Methodist HospitalHoustonTexasUSA
- Department of Radiation OncologyHouston Methodist HospitalHoustonTexasUSA
| |
Collapse
|
48
|
Xu Z, Liu T, Jiang Y, Chen Z, Shi X, Xu Y, Yu N, Hua X, Liang XJ, Yuan X, Guo S. Microcrystals of Ketal-Linked Paliperidone Prodrugs for Long-Acting Antipsychotics. Mol Pharm 2022; 19:3846-3857. [PMID: 36047719 DOI: 10.1021/acs.molpharmaceut.2c00348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Intramuscularly injectable long-acting prodrug-based microcrystals (MCs) are of particular interest for chronic disease management. Nevertheless, current prevalently used linkers degraded by enzymes have the potential drawback of substantial differences in enzyme levels between individuals. Here, we reported the synthesis of a stearyl-modified paliperidone prodrug (SKP) with an acid-sensitive ketal linker for developing long-acting MC antipsychotics. SKP-MCs of three different sizes were prepared and systematically examined. We found that paliperidone exposure in SKP-MC-treated rats was prolonged compared with that in rats treated with the commercial antipsychotic Invega Sustenna and that the drug release rate decreased with increasing MC size. In inflammation-inhibition-model rats, paliperidone release from the SKP-MCs was considerably decreased, indicating that the immune-mediated foreign-body response after intramuscular administration boosted paliperidone release. Our findings will provide valuable insights into in vivo drug release from prodrug-based MC formulations. The ketal-linked prodrug strategy might be a new solution for developing long-acting prodrug formulations of hydroxyl-group-bearing drugs.
Collapse
Affiliation(s)
- Zunkai Xu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Tao Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yaoyao Jiang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhixia Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoguang Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yang Xu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Na Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.,Translational Medicine Center, Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Xia Hua
- Aier Eye Institute, Changsha 410015, China.,Tianjin Aier Eye Hospital, Tianjin 300190, China
| | - Xing-Jie Liang
- Translational Medicine Center, Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China.,CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, China
| | - Xiaoyong Yuan
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, China.,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin 300020, China
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
49
|
Whyte W, Goswami D, Wang SX, Fan Y, Ward NA, Levey RE, Beatty R, Robinson ST, Sheppard D, O'Connor R, Monahan DS, Trask L, Mendez KL, Varela CE, Horvath MA, Wylie R, O'Dwyer J, Domingo-Lopez DA, Rothman AS, Duffy GP, Dolan EB, Roche ET. Dynamic actuation enhances transport and extends therapeutic lifespan in an implantable drug delivery platform. Nat Commun 2022; 13:4496. [PMID: 35922421 PMCID: PMC9349266 DOI: 10.1038/s41467-022-32147-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
Fibrous capsule (FC) formation, secondary to the foreign body response (FBR), impedes molecular transport and is detrimental to the long-term efficacy of implantable drug delivery devices, especially when tunable, temporal control is necessary. We report the development of an implantable mechanotherapeutic drug delivery platform to mitigate and overcome this host immune response using two distinct, yet synergistic soft robotic strategies. Firstly, daily intermittent actuation (cycling at 1 Hz for 5 minutes every 12 hours) preserves long-term, rapid delivery of a model drug (insulin) over 8 weeks of implantation, by mediating local immunomodulation of the cellular FBR and inducing multiphasic temporal FC changes. Secondly, actuation-mediated rapid release of therapy can enhance mass transport and therapeutic effect with tunable, temporal control. In a step towards clinical translation, we utilise a minimally invasive percutaneous approach to implant a scaled-up device in a human cadaveric model. Our soft actuatable platform has potential clinical utility for a variety of indications where transport is affected by fibrosis, such as the management of type 1 diabetes. Drug delivery implants suffer from diminished release profiles due to fibrous capsule formation over time. Here, the authors use soft robotic actuation to modulate the immune response of the host to maintain drug delivery over the longer-term and to perform controlled release in vivo.
Collapse
Affiliation(s)
- William Whyte
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Debkalpa Goswami
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sophie X Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yiling Fan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Niamh A Ward
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Ruth E Levey
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Rachel Beatty
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Scott T Robinson
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Declan Sheppard
- Department of Radiology, University Hospital, Galway, Ireland
| | - Raymond O'Connor
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - David S Monahan
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Lesley Trask
- Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Keegan L Mendez
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Claudia E Varela
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Markus A Horvath
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Robert Wylie
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Joanne O'Dwyer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Daniel A Domingo-Lopez
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Arielle S Rothman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| | - Eimear B Dolan
- Department of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland.
| | - Ellen T Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA.
| |
Collapse
|
50
|
Pan S, Upadhyay R, Singh S, Singh M. Synthesis, characterization, and functionalization of superadhesive melamine formaldehyde polyvinylpyrrolidone resins doped with metallic and graphene oxide nanoparticles for better industrial applications. J Appl Polym Sci 2022. [DOI: 10.1002/app.52921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sourav Pan
- School of Chemical Sciences Central University of Gujarat Gandhinagar India
| | - Richa Upadhyay
- School of Chemical Sciences Central University of Gujarat Gandhinagar India
| | - Sunita Singh
- Department of Biochemistry, Shivaji College, University of Delhi New Delhi India
| | - Man Singh
- School of Chemical Sciences Central University of Gujarat Gandhinagar India
| |
Collapse
|