151
|
Yao T, Baker MB, Moroni L. Strategies to Improve Nanofibrous Scaffolds for Vascular Tissue Engineering. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E887. [PMID: 32380699 PMCID: PMC7279151 DOI: 10.3390/nano10050887] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/19/2020] [Accepted: 04/24/2020] [Indexed: 12/25/2022]
Abstract
The biofabrication of biomimetic scaffolds for tissue engineering applications is a field in continuous expansion. Of particular interest, nanofibrous scaffolds can mimic the mechanical and structural properties (e.g., collagen fibers) of the natural extracellular matrix (ECM) and have shown high potential in tissue engineering and regenerative medicine. This review presents a general overview on nanofiber fabrication, with a specific focus on the design and application of electrospun nanofibrous scaffolds for vascular regeneration. The main nanofiber fabrication approaches, including self-assembly, thermally induced phase separation, and electrospinning are described. We also address nanofibrous scaffold design, including nanofiber structuring and surface functionalization, to improve scaffolds' properties. Scaffolds for vascular regeneration with enhanced functional properties, given by providing cells with structural or bioactive cues, are discussed. Finally, current in vivo evaluation strategies of these nanofibrous scaffolds are introduced as the final step, before their potential application in clinical vascular tissue engineering can be further assessed.
Collapse
Affiliation(s)
| | | | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands; (T.Y.); (M.B.B.)
| |
Collapse
|
152
|
Feng X, Xu P, Shen T, Zhang Y, Ye J, Gao C. Age-Related Regeneration of Osteochondral and Tibial Defects by a Fibrin-Based Construct in vivo. Front Bioeng Biotechnol 2020; 8:404. [PMID: 32432101 PMCID: PMC7214756 DOI: 10.3389/fbioe.2020.00404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/09/2020] [Indexed: 11/13/2022] Open
Abstract
Tissue-biomaterial interactions in different microenvironments influence significantly the repair and regeneration outcomes when a scaffold or construct is implanted. In order to elucidate this issue, a fibrin gel filled macroporous fibrin scaffold (fibrin-based scaffold) was fabricated by loading fibrinogen via a negative pressure method, following with thrombin crosslinking. The macroporous fibrin scaffold exhibited a porous structure with porosity of (88.1 ± 1.3)%, and achieved a modulus of 19.8 ± 0.4 kPa at a wet state after fibrin gel filling, providing a suitable microenvironment for bone marrow-derived mesenchymal stem cells (BMSCs). The in vitro cellular culture revealed that the fibrin-based scaffold could support the adhesion, spreading, and proliferation of BMSCs in appropriate cell encapsulation concentrations. The fibrin-based scaffolds were then combined with BMSCs and lipofectamine/plasmid deoxyribonucleic acid (DNA) encoding mouse-transforming growth factor β1 (pDNA-TGF-β1) complexes to obtain the fibrin-based constructs, which were implanted into osteochondral and tibial defects at young adult rabbits (3 months old) and aged adult rabbits (12 months old) to evaluate their respective repair effects. Partial repair of osteochondral defects and perfect restoration of tibial defects were realized at 18 weeks post-surgery for the young adult rabbits, whereas only partial repair of subchondral bone and tibial bone defects were found at the same time for the aged adult rabbits, confirming the adaptability of the fibrin-based constructs to the different tissue microenvironments by tissue-biomaterial interplays.
Collapse
Affiliation(s)
- Xue Feng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Peifang Xu
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Tao Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yihan Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Juan Ye
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
153
|
Mandal A, Clegg JR, Anselmo AC, Mitragotri S. Hydrogels in the clinic. Bioeng Transl Med 2020; 5:e10158. [PMID: 32440563 PMCID: PMC7237140 DOI: 10.1002/btm2.10158] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/21/2020] [Accepted: 03/22/2020] [Indexed: 12/13/2022] Open
Abstract
Injectable hydrogels are one of the most widely investigated and versatile technologies for drug delivery and tissue engineering applications. Hydrogels' versatility arises from their tunable structure, which has been enabled by considerable advances in fields such as materials engineering, polymer science, and chemistry. Advances in these fields continue to lead to invention of new polymers, new approaches to crosslink polymers, new strategies to fabricate hydrogels, and new applications arising from hydrogels for improving healthcare. Various hydrogel technologies have received regulatory approval for healthcare applications ranging from cancer treatment to aesthetic corrections to spinal fusion. Beyond these applications, hydrogels are being studied in clinical settings for tissue regeneration, incontinence, and other applications. Here, we analyze the current clinical landscape of injectable hydrogel technologies, including hydrogels that have been clinically approved or are currently being investigated in clinical settings. We summarize our analysis to highlight key clinical areas that hydrogels have found sustained success in and further discuss challenges that may limit their future clinical translation.
Collapse
Affiliation(s)
- Abhirup Mandal
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityCambridgeMassachusettsUSA
| | - John R. Clegg
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityCambridgeMassachusettsUSA
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityCambridgeMassachusettsUSA
| |
Collapse
|
154
|
Yan H, Hjorth M, Winkeljann B, Dobryden I, Lieleg O, Crouzier T. Glyco-Modification of Mucin Hydrogels to Investigate Their Immune Activity. ACS APPLIED MATERIALS & INTERFACES 2020; 12:19324-19336. [PMID: 32301325 PMCID: PMC7304668 DOI: 10.1021/acsami.0c03645] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/03/2020] [Indexed: 05/07/2023]
Abstract
Mucins are multifunctional glycosylated proteins that are increasingly investigated as building blocks of novel biomaterials. An attractive feature is their ability to modulate the immune response, in part by engaging with sialic acid binding receptors on immune cells. Once assembled into hydrogels, bovine submaxillary mucins (Muc gels) were shown to modulate the recruitment and activation of immune cells and avoid fibrous encapsulation in vivo. However, nothing is known about the early immune response to Muc gels. This study characterizes the response of macrophages, important orchestrators of the material-mediated immune response, over the first 7 days in contact with Muc gels. The role of mucin-bound sialic acid sugar residues was investigated by first enzymatically cleaving the sugar and then assembling the mucin variants into covalently cross-linked hydrogels with rheological and surface nanomechanical properties similar to nonmodified Muc gels. Results with THP-1 and human primary peripheral blood monocytes derived macrophages showed that Muc gels transiently activate the expression of both pro-inflammatory and anti-inflammatory cytokines and cell surface markers, for most makers with a maximum on the first day and loss of the effect after 7 days. The activation was sialic acid-dependent for a majority of the markers followed. The pattern of gene expression, protein expression, and functional measurements did not strictly correspond to M1 or M2 macrophage phenotypes. This study highlights the complex early events in macrophage activation in contact with mucin materials and the importance of sialic acid residues in such a response. The enzymatic glyco-modulation of Muc gels appears as a useful tool to help understand the biological functions of specific glycans on mucins which can further inform on their use in various biomedical applications.
Collapse
Affiliation(s)
- Hongji Yan
- Division of Glycoscience,
Department of Chemistry, School of Engineering Sciences in Chemistry,
Biotechnology and Health, KTH, Royal Institute
of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Morgan Hjorth
- Division of Glycoscience,
Department of Chemistry, School of Engineering Sciences in Chemistry,
Biotechnology and Health, KTH, Royal Institute
of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| | - Benjamin Winkeljann
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Boltzmannstrasse 11, 85748 Garching, Germany
| | - Illia Dobryden
- Division of Surface and Corrosion Science, Department of Chemistry,
School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Drottning Kristinas väg 51, 10044 Stockholm, Sweden
| | - Oliver Lieleg
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Boltzmannstrasse 11, 85748 Garching, Germany
| | - Thomas Crouzier
- Division of Glycoscience,
Department of Chemistry, School of Engineering Sciences in Chemistry,
Biotechnology and Health, KTH, Royal Institute
of Technology, AlbaNova University Center, 106 91 Stockholm, Sweden
| |
Collapse
|
155
|
Eslami-Kaliji F, Sarafbidabad M, Rajadas J, Mohammadi MR. Dendritic Cells as Targets for Biomaterial-Based Immunomodulation. ACS Biomater Sci Eng 2020; 6:2726-2739. [PMID: 33463292 DOI: 10.1021/acsbiomaterials.9b01987] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Various subtypes of immunocytes react against implanted biomaterials to eliminate the foreign body object from the host's body. Among these cells, dendritic cells (DCs) play a key role in early immune response, later engaging lymphocytes through antigens presentation. Due to their capability to induce tolerogenic or immunogenic responses, DCs have been considered as key therapeutic targets for immunomodulatory products. For instance, tolerogenic DCs are applied in the treatment of autoimmune diseases, rejection of allograft transplantation, and implanted biomaterial. Due to the emerging importance of DCs in immunomodulatory biomaterials, this Review summarizes DCs' responses-such as adhesion, migration, and maturation-to biomaterials. We also review some examples of key molecules and their applications in DCs' immunoengineering. These evaluations would pave the way for designing advanced biomaterials and nanomaterials to modulate the immune system, applicable in tissue engineering, transplantation, and drug delivery technologies.
Collapse
Affiliation(s)
- Farshid Eslami-Kaliji
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan 81746-73441, Iran
| | - Mohsen Sarafbidabad
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan 81746-73441, Iran
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, California 94305, United States.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco School of Pharmacy, San Francisco, California 94158, United States
| | - M Rezaa Mohammadi
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
156
|
Jimi S, Jaguparov A, Nurkesh A, Sultankulov B, Saparov A. Sequential Delivery of Cryogel Released Growth Factors and Cytokines Accelerates Wound Healing and Improves Tissue Regeneration. Front Bioeng Biotechnol 2020; 8:345. [PMID: 32426341 PMCID: PMC7212449 DOI: 10.3389/fbioe.2020.00345] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Growth factors and cytokines that are secreted by cells play a crucial role in the complex physiological reaction to tissue injury. The ability to spatially and temporally control their actions to maximize regenerative benefits and minimize side effects will help accelerate wound healing and improve tissue regeneration. In this study, the sequential targeted delivery of growth factor/cytokine combinations with regulatory functions on inflammation and tissue regeneration was examined using an internal splint wound healing model. Four examined growth factors and cytokines were effectively incorporated into a novel chitosan-based cryogel, which offered a controlled and sustained release of all factors while maintaining their biological activities. The cryogels incorporated with inflammation modulatory factors (IL-10 and TGF-β) and with wound healing factors (VEGF and FGF) were placed on the wound surface on day 0 and day 3, respectively, after wound initiation. Although wound area gradually decreased in all groups over time, the area in the cryogel group with growth factor/cytokine combinations was significantly reduced starting on day 7 and reached about 10% on day 10, as compared to 60-65% in the control groups. Sequential delivery of inflammation modulatory and wound healing factors enhanced granulation tissue formation, as well as functional neovascularization, leading to regenerative epithelialization. Collectively, the chitosan-based cryogel can serve as a controlled release system for sequential delivery of several growth factors and cytokines to accelerate tissue repair and regeneration.
Collapse
Affiliation(s)
- Shiro Jimi
- Central Laboratory for Pathology and Morphology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Alexandr Jaguparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Ayan Nurkesh
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bolat Sultankulov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
157
|
Schoenenberger AD, Tempfer H, Lehner C, Egloff J, Mauracher M, Bird A, Widmer J, Maniura-Weber K, Fucentese SF, Traweger A, Silvan U, Snedeker JG. Macromechanics and polycaprolactone fiber organization drive macrophage polarization and regulate inflammatory activation of tendon in vitro and in vivo. Biomaterials 2020; 249:120034. [PMID: 32315865 DOI: 10.1016/j.biomaterials.2020.120034] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/16/2022]
Abstract
Appropriate macrophage response to an implanted biomaterial is crucial for successful tissue healing outcomes. In this work we investigated how intrinsic topological cues from electrospun biomaterials and extrinsic mechanical loads cooperate to guide macrophage activation and macrophage-tendon fibroblast cross-talk. We performed a series of in vitro and in vivo experiments using aligned or randomly oriented polycaprolactone nanofiber substrates in both mechanically loaded and unloaded conditions. Across all experiments a disorganized biomaterial fiber topography was alone sufficient to promote a pro-inflammatory signature in macrophages, tendon fibroblasts, and tendon tissue. Extrinsic mechanical loading was found to strongly regulate the character of this signature by reducing pro-inflammatory markers both in vitro and in vivo. We observed that macrophages generally displayed a stronger response to biophysical cues than tendon fibroblasts, with dominant effects of cross-talk between these cell types observed in mechanical co-culture models. Collectively our data suggest that macrophages play a potentially important role as mechanosensory cells in tendon repair, and provide insight into how biological response might be therapeutically modulated by rational biomaterial designs that address the biomechanical niche of recruited cells.
Collapse
Affiliation(s)
- Angelina D Schoenenberger
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Herbert Tempfer
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University - Spinal Cord Injury & Tissue Regeneration Center Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christine Lehner
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University - Spinal Cord Injury & Tissue Regeneration Center Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Jasmin Egloff
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Marita Mauracher
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Anna Bird
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Jonas Widmer
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Katharina Maniura-Weber
- Biointerfaces, Empa, Swiss Federal Laboratories for Material Science and Technology, St. Gallen, Switzerland
| | - Sandro F Fucentese
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland
| | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University - Spinal Cord Injury & Tissue Regeneration Center Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Unai Silvan
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Jess G Snedeker
- Department of Orthopedics, Balgrist Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
158
|
Ferber S, Gonzalez RJ, Cryer AM, von Andrian UH, Artzi N. Immunology-Guided Biomaterial Design for Mucosal Cancer Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903847. [PMID: 31833592 DOI: 10.1002/adma.201903847] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/11/2019] [Indexed: 05/23/2023]
Abstract
Cancer of mucosal tissues is a major cause of worldwide mortality for which only palliative treatments are available for patients with late-stage disease. Engineered cancer vaccines offer a promising approach for inducing antitumor immunity. The route of vaccination plays a major role in dictating the migratory pattern of lymphocytes, and thus vaccine efficacy in mucosal tissues. Parenteral immunization, specifically subcutaneous and intramuscular, is the most common vaccination route. However, this induces marginal mucosal protection in the absence of tissue-specific imprinting signals. To circumvent this, the mucosal route can be utilized, however degradative mucosal barriers must be overcome. Hence, vaccine administration route and selection of materials able to surmount transport barriers are important considerations in mucosal cancer vaccine design. Here, an overview of mucosal immunity in the context of cancer and mucosal cancer clinical trials is provided. Key considerations are described regarding the design of biomaterial-based vaccines that will afford antitumor immune protection at mucosal surfaces, despite limited knowledge surrounding mucosal vaccination, particularly aided by biomaterials and mechanistic immune-material interactions. Finally, an outlook is given of how future biomaterial-based mucosal cancer vaccines will be shaped by new discoveries in mucosal vaccinology, tumor immunology, immuno-therapeutic screens, and material-immune system interplay.
Collapse
Affiliation(s)
- Shiran Ferber
- Department of Medicine, Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rodrigo J Gonzalez
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexander M Cryer
- Department of Medicine, Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Boston, MA, 02139, USA
| | - Natalie Artzi
- Department of Medicine, Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02139, USA
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| |
Collapse
|
159
|
Leppik L, Oliveira KMC, Bhavsar MB, Barker JH. Electrical stimulation in bone tissue engineering treatments. Eur J Trauma Emerg Surg 2020; 46:231-244. [PMID: 32078704 PMCID: PMC7113220 DOI: 10.1007/s00068-020-01324-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Electrical stimulation (EStim) has been shown to promote bone healing and regeneration both in animal experiments and clinical treatments. Therefore, incorporating EStim into promising new bone tissue engineering (BTE) therapies is a logical next step. The goal of current BTE research is to develop combinations of cells, scaffolds, and chemical and physical stimuli that optimize treatment outcomes. Recent studies demonstrating EStim's positive osteogenic effects at the cellular and molecular level provide intriguing clues to the underlying mechanisms by which it promotes bone healing. In this review, we discuss results of recent in vitro and in vivo research focused on using EStim to promote bone healing and regeneration and consider possible strategies for its application to improve outcomes in BTE treatments. Technical aspects of exposing cells and tissues to EStim in in vitro and in vivo model systems are also discussed.
Collapse
Affiliation(s)
- Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany.
| | - Karla Mychellyne Costa Oliveira
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - Mit Balvantray Bhavsar
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| | - John Howard Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, J.W. Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
160
|
Zhang J, Xie B, Xi Z, Zhao L, Cen L, Yang Y. A comparable study of polyglycolic acid's degradation on macrophages' activation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 109:110574. [DOI: 10.1016/j.msec.2019.110574] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/23/2019] [Accepted: 12/18/2019] [Indexed: 01/03/2023]
|
161
|
Duan Y, Zheng H, Li Z, Yao Y, Ding J, Wang X, Nakkala JR, Zhang D, Wang Z, Zuo X, Zheng X, Ling J, Gao C. Unsaturated polyurethane films grafted with enantiomeric polylysine promotes macrophage polarization to a M2 phenotype through PI3K/Akt1/mTOR axis. Biomaterials 2020; 246:120012. [PMID: 32276198 DOI: 10.1016/j.biomaterials.2020.120012] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022]
Abstract
The immune system responds immediately to tissue trauma and to biomaterial implants under the participation of M1/M2 macrophages polarization. The surface properties of biomaterials can significantly influence the tissue repair progress through modulating the macrophage functions. In this study, the surface of poly(propylene fumarate) polyurethane films (PPFU) is grafted with a same density of enantiomeric poly-l-lysine (PPFU-g-PLL) and poly-d-lysine (PPFU-g-PDL), leading to a similar level of enhanced surface wettability for the PPFU-g-PLL and PPFU-g-PDL. The polylysine-grafted PPFU can restrict the M1 polarization, whereas promote M2 polarization of macrophages in vitro, judging from the secretion of cytokines and expression of key M1 and M2 related genes. Comparatively, the PPFU-g-PDL has a stronger effect in inducing M2 polarization in vivo, resulting in a thinner fibrous capsule surrounding the implant biomaterials. The CD44 and integrins of macrophages participate in the polarization process probably by activating focal adhesion kinase (FAK) and Rho-associated protein kinase (ROCK), and downstream PI3K/Akt1/mTOR signal axis to up regulate M2 related gene expression. This study confirms for the first time that polylysine coating is an effective method to regulate the immune response of biomaterials, and the polylysine-modified thermoplastic PPFU with the advantage to promote M2 polarization may be applied widely in regenerative medicine.
Collapse
Affiliation(s)
- Yiyuan Duan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Honghao Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zehua Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xuemei Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jayachandra Reddy Nakkala
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Deteng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhaoyi Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xingang Zuo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiaowen Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jun Ling
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
162
|
Mukherjee S, Darzi S, Paul K, Cousins FL, Werkmeister JA, Gargett CE. Electrospun Nanofiber Meshes With Endometrial MSCs Modulate Foreign Body Response by Increased Angiogenesis, Matrix Synthesis, and Anti-Inflammatory Gene Expression in Mice: Implication in Pelvic Floor. Front Pharmacol 2020; 11:353. [PMID: 32265721 PMCID: PMC7107042 DOI: 10.3389/fphar.2020.00353] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/09/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Transvaginal meshes for the treatment of Pelvic Organ Prolapse (POP) have been associated with severe adverse events and have been banned for clinical use in many countries. We recently reported the design of degradable poly L-lactic acid-co-poly ε-caprolactone nanofibrous mesh (P nanomesh) bioengineered with endometrial mesenchymal stem/stromal cells (eMSC) for POP repair. We showed that such bioengineered meshes had high tissue integration as well as immunomodulatory effects in vivo. This study aimed to determine the key molecular players enabling eMSC-based foreign body response modulation. Methods SUSD2+ eMSC were purified from single cell suspensions obtained from endometrial biopsies from cycling women by magnetic bead sorting. Electrospun P nanomeshes with and without eMSC were implanted in a NSG mouse skin wound repair model for 1 and 6 weeks. Quantitative PCR was used to assess the expression of extracellular matrix (ECM), cell adhesion, angiogenesis and inflammation genes as log2 fold changes compared to sham controls. Histology and immunostaining were used to visualize the ECM, blood vessels, and multinucleated foreign body giant cells around implants. Results Bioengineered P nanomesh/eMSC constructs explanted after 6 weeks showed significant increase in 35 genes associated with ECM, ECM regulation, cell adhesion angiogenesis, and immune response in comparison to P nanomesh alone. In the absence of eMSC, acute inflammatory genes were significantly elevated at 1 week. However, in the presence of eMSC, there was an increased expression of anti-inflammatory genes including Mrc1 and Arg1 by 6 weeks. There was formation of multinucleated foreign body giant cells around both implants at 6 weeks that expressed CD206, a M2 macrophage marker. Conclusion This study reveals that eMSC modulate the foreign body response to degradable P nanomeshes in vivo by altering the expression profile of mouse genes. eMSC reduce acute inflammatory and increase ECM synthesis, angiogenesis and anti-inflammatory gene expression at 6 weeks while forming newly synthesized collagen within the nanomeshes and neo-vasculature in close proximity. From a tissue engineering perspective, this is a hallmark of a highly successful implant, suggesting significant potential as alternative surgical constructs for the treatment of POP.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Fiona L Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
163
|
Liu G, Fu M, Li F, Fu W, Zhao Z, Xia H, Niu Y. Tissue-engineered PLLA/gelatine nanofibrous scaffold promoting the phenotypic expression of epithelial and smooth muscle cells for urethral reconstruction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110810. [PMID: 32279818 DOI: 10.1016/j.msec.2020.110810] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 01/31/2023]
Abstract
The repair and regeneration of tissues using tissue-engineered scaffolds represent the ultimate goal of regenerative medicine. Despite rapid developments in the field, urethral tissue engineering methods are still insufficient to replicate natural urethral tissue because the bioactivity of existing scaffolds is inefficient, especially for large tissue defects, which require large tissue-engineered scaffolds. Here, we describe the efficiency of gelatine-functionalized, tubular nanofibrous scaffolds of poly(l-lactic acid) (PLLA) in regulating the phenotypic expression of epithelial cells (ECs) and smooth muscle cells (SMCs) for urethral reconstruction. Flexible PLLA/gelatine tubular nanofibrous scaffolds with hierarchical architecture were fabricated by electrospinning. The PLLA/gelatine nanofibrous scaffold exhibited enhanced hydrophilicity and significantly promoted the adhesion, oriented elongation, and proliferation of New Zealand rabbit autologous ECs and SMCs simultaneously. Compared with pure PLLA nanofibrous scaffold, PLLA/gelatine nanofibrous scaffolds upregulated the expression of keratin (AE1/AE3) in ECs and actin (α-SMA) in SMCs as well as the synthesis of elastin. Three months of in vivo scaffold replacement of New Zealand rabbit urethras indicated that a tubular cellularized PLLA/gelatine nanofibrous scaffold maintained urethral patency and facilitated oriented SMC remodeling, lumen epithelialization, and angiogenesis. Our observations showed the synergistic effects of nano-morphology and biochemical clues in the design of biomimetic scaffolds, which can effectively promote urethral regeneration.
Collapse
Affiliation(s)
- Guochang Liu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Ming Fu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Feng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Wen Fu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Zhang Zhao
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China.
| | - Yuqing Niu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
164
|
Zhao Z, Zhao Q, Gu B, Yin C, Shen K, Tang H, Xia H, Zhang X, Zhao Y, Yang X, Zhang Y. Minimally invasive implantation and decreased inflammation reduce osteoinduction of biomaterial. Am J Cancer Res 2020; 10:3533-3545. [PMID: 32206106 PMCID: PMC7069090 DOI: 10.7150/thno.39507] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/28/2020] [Indexed: 02/06/2023] Open
Abstract
Surgical trauma of biomaterial implantation significantly influences the immune system and the biological effects of biomaterials. Minimally invasive surgery has become a trend of clinical development but violating the concept of osteoimmunomodulation will hinder the biological effects of materials. Our study focused on biphasic calcium phosphate (BCP), the ectopia osteoinductive materials, filling the research blank of the significance of adaptive immunity crosstalk with bone biomaterials, and improving the interaction mechanism between bone biomaterials and immune response. Methods: The BCP bioceramics were implanted by conventional and minimally invasive methods in the gastrocnemius wild-type or T cells depleted mice to test the effect of ectopia osteoinduction. Moreover, flow cytometry was used to detect immune responses, T cell sorting and Western Blot molecular biology experiments, and transwell assays migration of mesenchymal stem cells (MSCs). Results: We found that BCP, an implantable osteoinductive material, could not activate the adaptive immune response mediated by T cells after minimally invasive surgery. Further studies revealed that under the conventional non-minimally invasive BCP implantation, a positive correlation existed between T cell recruitment and the infiltration and osteogenic differentiation of MSCs. Interestingly, after BCP was implanted by minimally invasive surgery or implanted in T cell depleted mice, MSCs infiltration and osteogenic differentiation were significantly reduced, and BCP could not achieve the biological effects of ectopia ossification. Finally, we confirmed that a certain extent inflammatory stimulation activated the adaptive immune response mediated by T cells, up-regulated the nuclear factor-κB (NF-κB) signal in T cells, released a large amount of chemokine C-C motif chemokine ligand 5(CCL5) to recruit MSCs to the surrounding material, and finally achieved the ideal effect of osteoinduction. Conclusion: From experimental research and clinical surgery, this study discovered that the T cells are indispensable in the ectopia ossification mediated by osteoinductive materials, put forward and confirmed the surgery method as a key variable factor restricting the application effect of biological materials, enriched the key mechanism of adaptive immunity in osteoimmunomodulation, and laid a theoretical foundation for the development of osteoinductive materials and bone tissue regeneration.
Collapse
|
165
|
Umuhoza D, Yang F, Long D, Hao Z, Dai J, Zhao A. Strategies for Tuning the Biodegradation of Silk Fibroin-Based Materials for Tissue Engineering Applications. ACS Biomater Sci Eng 2020; 6:1290-1310. [DOI: 10.1021/acsbiomaterials.9b01781] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Diane Umuhoza
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
- Commercial Insect Program, Sericulture, Rwanda Agricultural Board, 5016 Kigali, Rwanda
| | - Fang Yang
- Department of Biomaterials, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Dingpei Long
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| | - Zhanzhang Hao
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| | - Jing Dai
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| | - Aichun Zhao
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400716, People’s Republic of China
| |
Collapse
|
166
|
Cui Z, Lin J, Zhan C, Wu J, Shen S, Si J, Wang Q. Biomimetic composite scaffolds based on surface modification of polydopamine on ultrasonication induced cellulose nanofibrils (CNF) adsorbing onto electrospun thermoplastic polyurethane (TPU) nanofibers. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:561-577. [DOI: 10.1080/09205063.2019.1705534] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Zhixiang Cui
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| | - Jixin Lin
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| | - Conghua Zhan
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| | - Jiahui Wu
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| | - Shuai Shen
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| | - Junhui Si
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| | - Qianting Wang
- School of Materials Science and Engineering, Fujian University of Technology, Fujian, China
- Fujian Provincial Key Laboratory of Advanced Materials Processing and Application, Fujian, China
- Fujian Provincial Engineering Research Center of Die & Mold, Fujian University of Technology, Fujian, China
| |
Collapse
|
167
|
Yoshimoto Y, Jo JI, Tabata Y. Preparation of antibody-immobilized gelatin nanospheres incorporating a molecular beacon to visualize the biological function of macrophages. Regen Ther 2020; 14:11-18. [PMID: 31970268 PMCID: PMC6961756 DOI: 10.1016/j.reth.2019.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/15/2019] [Accepted: 12/24/2019] [Indexed: 12/22/2022] Open
Abstract
Introduction Inflammatory response plays an important role in the disease progress or therapeutic effect. In this context, it is highly required to develop a technology to visualize the inflammatory response. In this study, macrophages and their microRNA (miRNA) which are involved in the inflammatory response, were focused while a system of molecular beacon (MB) to detect the miRNA of macrophages was designed and prepared. Methods Gelatin nanospheres were prepared by the conventional coacervation method. An antibody with an affinity for the surface receptor of macrophages was immobilized onto the gelatin nanospheres by several methods. A nucleic acid-based MB for a pro-inflammatory miRNA 155–5p was designed and incorporated into the antibody-immobilized gelatin nanospheres (MB-gelatin NS). Macrophages before and after the polarization into pro-inflammatory or anti-inflammatory phenotypes were cultured with the MB-gelatin NS and change in the intracellular fluorescence was observed. Results The antibody-immobilized gelatin nanospheres prepared by a coupling between the amino groups of gelatin and the sugar chains of antibody with NaIO4 showed the highest affinity for cellular receptor. MB complexed with the cell-penetrating (CP) peptide was successfully incorporated into the antibody-immobilized gelatin nanospheres. When cultured with pro-inflammatory macrophages, MB-gelatin NS efficiently detected the miRNA 155–5p to emit fluorescence. Conclusions By the NaIO4 method, the antibody was immobilized onto gelatin nanospheres with a high affinity remaining while the MB was incorporated into the antibody-immobilized gelatin nanospheres. The MB incorporated allowed mRNA to visualize the pro-inflammatory nature of macrophages. Antibody could be immobilized onto gelatin nanospheres with the affinity remaining. MB for a pro-inflammatory miRNA was incorporated into gelatin nanospheres. MB incorporated emitted the fluorescence in the pro-inflammatory macrophages.
Collapse
Key Words
- Antibody immobilization
- BCA, bicinchoninic acid
- BHQ, black hole quencher
- BSA, bovine serum albumin
- CP, cell-penetrating
- DDW, double-distilled water
- DLS, dynamic light scattering
- DSS, disuccinimidyl suberate
- FCS, fetal calf serum
- GA, glutaraldehyde
- Gelatin nanospheres
- IL, interleukin
- Ig, immunoglobulin
- Inflammatory response
- KPB, potassium phosphate-buffered
- MB, molecular beacon
- Macrophages
- Molecular beacon
- PBS, phosphate buffered-saline
- WST-8, 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium
- miRNA, microRNA
- microRNA
- qRT-PCR, quantitative real time-polymerase chain reaction
Collapse
Affiliation(s)
- Yu Yoshimoto
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
168
|
Sohutskay DO, Puls TJ, Voytik-Harbin SL. Collagen Self-assembly: Biophysics and Biosignaling for Advanced Tissue Generation. MULTI-SCALE EXTRACELLULAR MATRIX MECHANICS AND MECHANOBIOLOGY 2020. [DOI: 10.1007/978-3-030-20182-1_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
169
|
Zakeri Siavashani A, Mohammadi J, Maniura-Weber K, Senturk B, Nourmohammadi J, Sadeghi B, Huber L, Rottmar M. Silk based scaffolds with immunomodulatory capacity: anti-inflammatory effects of nicotinic acid. Biomater Sci 2020; 8:148-162. [DOI: 10.1039/c9bm00814d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Here we show that 3D silk scaffolds loaded with nicotinic acid have great potential for tissue engineering due to their excellent cytocompatibility and ability to decrease the expression of proinflammatory markers in a concentration dependent manner.
Collapse
Affiliation(s)
| | - Javad Mohammadi
- Faculty of New Sciences and Technologies
- University of Tehran
- Tehran
- Iran
| | - Katharina Maniura-Weber
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Biointerfaces
- St.Gallen
- Switzerland
| | - Berna Senturk
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Biointerfaces
- St.Gallen
- Switzerland
| | | | - Behnam Sadeghi
- Translational Cell therapy Research (TCR)
- Department of CLINTEC
- Karolinska Institutet
- Stockholm
- Sweden
| | - Lukas Huber
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Laboratory for Building Energy Materials and Components
- Dübendorf
- Switzerland
| | - Markus Rottmar
- Empa
- Swiss Federal Laboratories for Materials Science and Technology
- Biointerfaces
- St.Gallen
- Switzerland
| |
Collapse
|
170
|
Feng X, Xu P, Shen T, Zhang Y, Ye J, Gao C. Influence of pore architectures of silk fibroin/collagen composite scaffolds on the regeneration of osteochondral defects in vivo. J Mater Chem B 2020; 8:391-405. [DOI: 10.1039/c9tb01558b] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aligned scaffolds facilitate migration of endogenous reparative cells, leading to better regeneration of osteochondral defects.
Collapse
Affiliation(s)
- Xue Feng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Peifang Xu
- Department of Ophthalmology
- The Second Affiliated Hospital of Zhejiang University
- College of Medicine
- Hangzhou
- P. R. China
| | - Tao Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Yihan Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University
- Hangzhou 310027
- P. R. China
| | - Juan Ye
- Department of Ophthalmology
- The Second Affiliated Hospital of Zhejiang University
- College of Medicine
- Hangzhou
- P. R. China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University
- Hangzhou 310027
- P. R. China
| |
Collapse
|
171
|
Zhang J, Song C, Han Y, Xi Z, Zhao L, Cen L, Yang Y. Regulation of inflammatory response to polyglycolic acid scaffolds through incorporation of sodium tripolyphosphate. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2019.109349] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
172
|
Yang D, Xiao J, Wang B, Li L, Kong X, Liao J. The immune reaction and degradation fate of scaffold in cartilage/bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109927. [DOI: 10.1016/j.msec.2019.109927] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023]
|
173
|
Zhu M, Li W, Dong X, Yuan X, Midgley AC, Chang H, Wang Y, Wang H, Wang K, Ma PX, Wang H, Kong D. In vivo engineered extracellular matrix scaffolds with instructive niches for oriented tissue regeneration. Nat Commun 2019; 10:4620. [PMID: 31604958 PMCID: PMC6789018 DOI: 10.1038/s41467-019-12545-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/12/2019] [Indexed: 12/22/2022] Open
Abstract
Implanted scaffolds with inductive niches can facilitate the recruitment and differentiation of host cells, thereby enhancing endogenous tissue regeneration. Extracellular matrix (ECM) scaffolds derived from cultured cells or natural tissues exhibit superior biocompatibility and trigger favourable immune responses. However, the lack of hierarchical porous structure fails to provide cells with guidance cues for directional migration and spatial organization, and consequently limit the morpho-functional integration for oriented tissues. Here, we engineer ECM scaffolds with parallel microchannels (ECM-C) by subcutaneous implantation of sacrificial templates, followed by template removal and decellularization. The advantages of such ECM-C scaffolds are evidenced by close regulation of in vitro cell activities, and enhanced cell infiltration and vascularization upon in vivo implantation. We demonstrate the versatility and flexibility of these scaffolds by regenerating vascularized and innervated neo-muscle, vascularized neo-nerve and pulsatile neo-artery with functional integration. This strategy has potential to yield inducible biomaterials with applications across tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Meifeng Zhu
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
- Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Wen Li
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
| | - Xianhao Dong
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
| | - Xingyu Yuan
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
| | - Adam C Midgley
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
| | - Hong Chang
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
| | - Yuhao Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Haoyu Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Kai Wang
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China.
| | - Peter X Ma
- Department of Biologic and Materials Sciences, Department of Biomedical Engineering, Macromolecular Science and Engineering Centre, Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA.
| | - Deling Kong
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China.
- Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
174
|
Yang C, Zhao C, Wang X, Shi M, Zhu Y, Jing L, Wu C, Chang J. Stimulation of osteogenesis and angiogenesis by micro/nano hierarchical hydroxyapatite via macrophage immunomodulation. NANOSCALE 2019; 11:17699-17708. [PMID: 31545331 DOI: 10.1039/c9nr05730g] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Biomaterial topography-based strategies are regarded as an effective way to regulate the osteoimmune environment which plays an indispensable role in the bone regeneration process. The rapid development of manufacture techniques makes it possible to investigate the cell-topography interactions by preparing various micro and nano-topographical surfaces on biomaterials. Still, it is a challenge to prepare well-defined micro/nano hierarchical structures of bioceramics due to the inherent brittleness of ceramic materials. Also, the correlation between osteoimmunomodulation initiated by micro/nano hierarchical topographies and the tissue regeneration outcomes is unclear. In this study, we prepared well-defined micro/nano hierarchical structures on hydroxyapatite (HA) bioceramics through the combination of the photolithography and hydrothermal techniques. Three different microscale circular patterns (4 μm, 12 μm and 36 μm) and nanotopographies (nanoneedle, nanosheet and nanorod) were fabricated by changing the size of the mask and the condition of the hydrothermal reaction. The macrophage responses on the nanoneedle structures with different micropatterns were investigated and the micro/nano hierarchical structures with appropriate pattern sizes could either promote or alleviate the macrophage polarization, which further affected the outcomes of the osteogenic differentiation of human bone marrow stromal cells (hBMSCs) and angiogenic activity of human umbilical vein endothelial cells (HUVECs). Our study demonstrated that osteoimmunomodulation could be manipulated via tuning the micro/nano hierarchical structures, which could lead to a new strategy for the development of bone biomaterials with favorable osteoimmunomodulatory properties.
Collapse
Affiliation(s)
- Chen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China.
| | | | | | | | | | | | | | | |
Collapse
|
175
|
Kilic Bektas C, Burcu A, Gedikoglu G, Telek HH, Ornek F, Hasirci V. Methacrylated gelatin hydrogels as corneal stroma substitutes: in vivo study. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 30:1803-1821. [DOI: 10.1080/09205063.2019.1666236] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Cemile Kilic Bektas
- Departments of Biological Sciences, Middle East Technical University (METU), Ankara, Turkey
- Department of Biotechnology, Middle East Technical University (METU), Ankara, Turkey
- BIOMATEN, METU Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey
| | - Ayse Burcu
- Eye Clinic, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Gokhan Gedikoglu
- Department of Medical Pathology, Hacettepe University, Ankara, Turkey
| | - Hande H. Telek
- Eye Clinic, Beytepe Murat Erdi Eker State Hospital, Ankara, Turkey
| | - Firdevs Ornek
- Eye Clinic, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Vasif Hasirci
- Departments of Biological Sciences, Middle East Technical University (METU), Ankara, Turkey
- Department of Biotechnology, Middle East Technical University (METU), Ankara, Turkey
- BIOMATEN, METU Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey
- Department of Medical Engineering, Acıbadem Mehmet Ali Aydınlar University, İstanbul, Turkey
| |
Collapse
|
176
|
Artsen AM, Rytel M, Liang R, King GE, Meyn L, Abramowitch SD, Moalli PA. Mesh induced fibrosis: The protective role of T regulatory cells. Acta Biomater 2019; 96:203-210. [PMID: 31326666 DOI: 10.1016/j.actbio.2019.07.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023]
Abstract
Polypropylene mesh is widely used in urogynecologic surgery, but complications rates (pain and exposure) approach 10%. Emerging evidence implicates the adaptive immune system in regulating the foreign body response to mesh, particularly regulatory T cells (Tregs), which modify macrophage differentiation and down-regulate CD8+ effector T cells. We hypothesize that Tregs protect against a profibrotic response, a likely mechanism of pain complications. Here, thin sections of mesh-tissue complexes removed for the primary complaint of pain (N = 14) or exposure (N = 15) were labeled for CD8, CD4 (Th), and FoxP3 (Tregs) via immunofluorescence. The same sections were analyzed for localized collagen deposition via a customized semi-quantitative assessment (0.25 mm2 grid) after trichrome staining. TGF-β1 concentrations were determined by enzyme-linked immunosorbent assay. Fewer Treg and CD4+ cells were found in fibrotic areas versus non-fibrotic areas (503 and 550/cm2 fewer, respectively, both P < 0.001). TGF-β1 was higher in mesh samples compared to autologous control biopsies. TGF-β 1 inversely correlated with age, r -0.636(p = 0.008). No differences were found in T cell subgroups or fibrotic indices between pain and exposure groups. A moderate inverse relationship was found between TGF-β1 and Tregs (r -0.402, P = 0.009). Tregs were present up to 12 years after mesh implantation, challenging the assumption that the adaptive immune response to a foreign body is transient. In conclusion, the inverse relationship between fibrosis and Tregs, and TGF-β1 and Tregs points to a protective role of these cells. Similar immunologic responses in patients with pain and exposure suggest these complications exist along a spectrum. STATEMENT OF SIGNIFICANCE: The use of polypropylene mesh has been associated with improved outcomes in urogynecologic surgery, but is associated with significant complications, including pain and exposure through the vaginal epithelium. The host immune response features a prolonged inflammatory reaction containing innate immune cells and T lymphocytes clustered in capsules around the mesh fibers. This study uncovers the inverse relationship between T regulatory cells and the extent of fibrosis around the mesh, suggesting an anti-fibrotic effect. In addition, concentrations of T regulatory and T effector cells and levels of fibrosis connect these two most common complications into one mechanistic pathway. These new insights into the immune response to implanted mesh are an important step in understanding the causes of these surgical complications.
Collapse
|
177
|
Sapru S, Das S, Mandal M, Ghosh AK, Kundu SC. Nonmulberry silk protein sericin blend hydrogels for skin tissue regeneration - in vitro and in vivo. Int J Biol Macromol 2019; 137:545-553. [DOI: 10.1016/j.ijbiomac.2019.06.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/15/2019] [Accepted: 06/17/2019] [Indexed: 12/30/2022]
|
178
|
Jafarkhani M, Salehi Z, Bagheri Z, Aayanifard Z, Rezvan A, Doosthosseini H, Shokrgozar MA. Graphene functionalized decellularized scaffold promotes skin cell proliferation. CAN J CHEM ENG 2019. [DOI: 10.1002/cjce.23588] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mahboubeh Jafarkhani
- School of Chemical EngineeringCollege of Engineering, University of Tehran Iran
- DTU NanotechTechnical University of Denmark, KongensLyngby Region Hovedstaden Denmark
| | - Zeinab Salehi
- School of Chemical EngineeringCollege of Engineering, University of Tehran Iran
| | - Zahra Bagheri
- School of Chemical EngineeringCollege of Engineering, University of Tehran Iran
| | - Zahra Aayanifard
- School of Chemical EngineeringCollege of Engineering, University of Tehran Iran
| | - Ali Rezvan
- School of Chemical EngineeringCollege of Engineering, University of Tehran Iran
| | - Hamid Doosthosseini
- School of Chemical EngineeringCollege of Engineering, University of Tehran Iran
| | | |
Collapse
|
179
|
Smith ES, Porterfield JE, Kannan RM. Leveraging the interplay of nanotechnology and neuroscience: Designing new avenues for treating central nervous system disorders. Adv Drug Deliv Rev 2019; 148:181-203. [PMID: 30844410 PMCID: PMC7043366 DOI: 10.1016/j.addr.2019.02.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022]
Abstract
Nanotechnology has the potential to open many novel diagnostic and treatment avenues for disorders of the central nervous system (CNS). In this review, we discuss recent developments in the applications of nanotechnology in CNS therapies, diagnosis and biology. Novel approaches for the diagnosis and treatment of neuroinflammation, brain dysfunction, psychiatric conditions, brain cancer, and nerve injury provide insights into the potential of nanomedicine. We also highlight nanotechnology-enabled neuroscience techniques such as electrophysiology and intracellular sampling to improve our understanding of the brain and its components. With nanotechnology integrally involved in the advancement of basic neuroscience and the development of novel treatments, combined diagnostic and therapeutic applications have begun to emerge. Nanotheranostics for the brain, able to achieve single-cell resolution, will hasten the rate in which we can diagnose, monitor, and treat diseases. Taken together, the recent advances highlighted in this review demonstrate the prospect for significant improvements to clinical diagnosis and treatment of a vast array of neurological diseases. However, it is apparent that a strong dialogue between the nanoscience and neuroscience communities will be critical for the development of successful nanotherapeutics that move to the clinic, benefit patients, and address unmet needs in CNS disorders.
Collapse
Affiliation(s)
- Elizabeth S Smith
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joshua E Porterfield
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA; Kennedy Krieger Institute, Johns Hopkins University for Cerebral Palsy Research Excellence, Baltimore, MD 21218, USA.
| |
Collapse
|
180
|
Sahle FF, Kim S, Niloy KK, Tahia F, Fili CV, Cooper E, Hamilton DJ, Lowe TL. Nanotechnology in regenerative ophthalmology. Adv Drug Deliv Rev 2019; 148:290-307. [PMID: 31707052 PMCID: PMC7474549 DOI: 10.1016/j.addr.2019.10.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/18/2022]
Abstract
In recent years, regenerative medicine is gaining momentum and is giving hopes for restoring function of diseased, damaged, and aged tissues and organs and nanotechnology is serving as a catalyst. In the ophthalmology field, various types of allogenic and autologous stem cells have been investigated to treat some ocular diseases due to age-related macular degeneration, glaucoma, retinitis pigmentosa, diabetic retinopathy, and corneal and lens traumas. Nanomaterials have been utilized directly as nanoscaffolds for these stem cells to promote their adhesion, proliferation and differentiation or indirectly as vectors for various genes, tissue growth factors, cytokines and immunosuppressants to facilitate cell reprogramming or ocular tissue regeneration. In this review, we reviewed various nanomaterials used for retina, cornea, and lens regenerations, and discussed the current status and future perspectives of nanotechnology in tracking cells in the eye and personalized regenerative ophthalmology. The purpose of this review is to provide comprehensive and timely insights on the emerging field of nanotechnology for ocular tissue engineering and regeneration.
Collapse
Affiliation(s)
- Fitsum Feleke Sahle
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Sangyoon Kim
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Kumar Kulldeep Niloy
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Faiza Tahia
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Cameron V Fili
- Department of Comparative Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Emily Cooper
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - David J Hamilton
- Department of Comparative Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Tao L Lowe
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA.
| |
Collapse
|
181
|
Das D, Noh I. Overviews of Biomimetic Medical Materials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1064:3-24. [PMID: 30471023 DOI: 10.1007/978-981-13-0445-3_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This chapter describes the overviews of biomimetic medical materials which covers innovation and significance of terminology, diverse fabrication methods, and technologies ranges from nanotechnology to 3D printing to develop biomimetic materials for medical applications. It also depicts specific fundamental characteristics required for a material to be a model biomimetic material for particular medical application. It basically outlines current statuses of biomimetic medical materials used for tissue engineering and regenerative medicine, drug/protein delivery, bioimaging, biosensing, and 3D bioprinting technology. It also illustrates the effect of functionalization of a material through chemical and biological approaches towards different applications. Not only, the key properties and potential applications of the biomimetic materials, but it also explains the protection and utilization of intellectual property associated with biomedical materials.
Collapse
Affiliation(s)
- Dipankar Das
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, South Korea.,Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, South Korea
| | - Insup Noh
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, South Korea. .,Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, South Korea.
| |
Collapse
|
182
|
Hachim D, Iftikhar A, LoPresti ST, Nolfi AL, Ravichandar S, Skillen CD, Brown BN. Distinct release strategies are required to modulate macrophage phenotype in young versus aged animals. J Control Release 2019; 305:65-74. [PMID: 31103676 PMCID: PMC6602858 DOI: 10.1016/j.jconrel.2019.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/09/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
The role of innate immunity and macrophages in the host response to biomaterials has received renewed attention. A context-dependent spectrum of macrophage phenotypes are shown to affect tissue integration and performance of implanted biomaterials and medical devices. Recent studies by our group demonstrated that the host response in aged animals was characterized by delayed macrophage recruitment, differences in marker expression and a shifted pro-inflammatory (M1) response, associated with an unresolved host response in the long-term. The present work sought to study the effects of single and sequential cytokine delivery regimens in aged mice to restore delayed recruitment of macrophages and shift the inflammatory host response towards an M2-like phenotype, using MCP-1 (macrophage chemotactic protein-1) and IL-4 (interleukin-4), respectively. Implantation of cytokine-eluting implants showed a preserved response to MCP-1 in both young and aged animals, restoring delayed macrophage recruitment in aged mice. However, the response elicited by IL-4, sequential delivery of MCP-1/IL-4 and coating components was distinct in young versus aged mice. While single delivery of IL-4 did not counteract the high inflammatory response observed in aged mice, the sequential delivery of MCP-1/IL-4 was capable of restoring both recruitment and shifting the macrophage response towards an M2-like phenotype, associated with decreased implant scarring in the long-term. In young mice, sequential delivery was not as effective as IL-4 alone at promoting an M2-like response, but did result in a reduction of M1 macrophages and capsule deposition downstream. These results demonstrate that a proper understanding of patient/context-dependent biological responses are needed to design biomaterial-based therapies with improved outcomes in the setting of aging.
Collapse
Affiliation(s)
- Daniel Hachim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States of America
| | - Aimon Iftikhar
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States of America
| | - Samuel T LoPresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States of America
| | - Alexis L Nolfi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States of America
| | - Shweta Ravichandar
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States of America
| | - Clint D Skillen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America
| | - Bryan N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States of America; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States of America; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA 15213, United States of America.
| |
Collapse
|
183
|
Acellular Biologic Scaffolds in Regenerative Medicine: Unacceptable Variability with Acceptable Results. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00106-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
184
|
Alagpulinsa DA, Cao JJL, Driscoll RK, Sîrbulescu RF, Penson MFE, Sremac M, Engquist EN, Brauns TA, Markmann JF, Melton DA, Poznansky MC. Alginate-microencapsulation of human stem cell-derived β cells with CXCL12 prolongs their survival and function in immunocompetent mice without systemic immunosuppression. Am J Transplant 2019; 19:1930-1940. [PMID: 30748094 DOI: 10.1111/ajt.15308] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 01/25/2023]
Abstract
Pancreatic β-cell replacement by islet transplantation for the treatment of type 1 diabetes (T1D) is currently limited by donor tissue scarcity and the requirement for lifelong immunosuppression. The advent of in vitro differentiation protocols for generating functional β-like cells from human pluripotent stem cells, also referred to as SC-β cells, could eliminate these obstacles. To avoid the need for immunosuppression, alginate-microencapsulation is widely investigated as a safe path to β-cell replacement. Nonetheless, inflammatory foreign body responses leading to pericapsular fibrotic overgrowth often causes microencapsulated islet-cell death and graft failure. Here we used a novel approach to evade the pericapsular fibrotic response to alginate-microencapsulated SC-β cells; an immunomodulatory chemokine, CXCL12, was incorporated into clinical grade sodium alginate to microencapsulate SC-β cells. CXCL12 enhanced glucose-stimulated insulin secretion activity of SC-β cells and induced expression of genes associated with β-cell function in vitro. SC-β cells co-encapsulated with CXCL12 showed enhanced insulin secretion in diabetic mice and accelerated the normalization of hyperglycemia. Additionally, SC-β cells co-encapsulated with CXCL12 evaded the pericapsular fibrotic response, resulting in long-term functional competence and glycemic correction (>150 days) without systemic immunosuppression in immunocompetent C57BL/6 mice. These findings lay the groundwork for further preclinical translation of this approach into large animal models of T1D.
Collapse
Affiliation(s)
- David A Alagpulinsa
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jenny J L Cao
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Pathology and School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Riley K Driscoll
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah
| | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Madeline F E Penson
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marinko Sremac
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Elise N Engquist
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Timothy A Brauns
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - James F Markmann
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
185
|
|
186
|
Fernández-Colino A, Iop L, Ventura Ferreira MS, Mela P. Fibrosis in tissue engineering and regenerative medicine: treat or trigger? Adv Drug Deliv Rev 2019; 146:17-36. [PMID: 31295523 DOI: 10.1016/j.addr.2019.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/11/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Fibrosis is a life-threatening pathological condition resulting from a dysfunctional tissue repair process. There is no efficient treatment and organ transplantation is in many cases the only therapeutic option. Here we review tissue engineering and regenerative medicine (TERM) approaches to address fibrosis in the cardiovascular system, the kidney, the lung and the liver. These strategies have great potential to achieve repair or replacement of diseased organs by cell- and material-based therapies. However, paradoxically, they might also trigger fibrosis. Cases of TERM interventions with adverse outcome are also included in this review. Furthermore, we emphasize the fact that, although organ engineering is still in its infancy, the advances in the field are leading to biomedically relevant in vitro models with tremendous potential for disease recapitulation and development of therapies. These human tissue models might have increased predictive power for human drug responses thereby reducing the need for animal testing.
Collapse
|
187
|
Preparation of fibrin hydrogels to promote the recruitment of anti-inflammatory macrophages. Acta Biomater 2019; 89:152-165. [PMID: 30862554 DOI: 10.1016/j.actbio.2019.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 12/31/2022]
Abstract
Macrophages play an important role in regulating inflammation and tissue regeneration. In the present study, uniform fibrin hydrogel scaffolds were engineered in millimeters. These scaffolds induced anti-inflammatory macrophages to digest and infiltrate the scaffold. The culture conditions of the fibrin hydrogels decreased the secretion of tumor necrosis factor-α (TNF-α), a pro-inflammatory cytokine, and increased the secretion of interleukin-10 (IL-10), an anti-inflammatory cytokine, in mouse bone marrow-derived macrophages. Similar results were also observed in human monocyte-derived macrophages (HMDMs). In addition, most of cells that infiltrated the fibrin hydrogels were macrophages expressing CD163, CD204, and CD206, which are anti-inflammatory macrophages markers, both in mice and in human cells. Therefore, to induce increased macrophage infiltration, we attempted to combine fibrin hydrogels with SEW2871, a monocyte/macrophage recruitment agent that is known to be a sphingosine-1 phosphate receptor 1 agonist, solubilized in water by micelle formation with a cholesterol-grafted gelatin. However, the fibrin hydrogels alone retained the same monocyte migration activity as the hydrogels with SEW2871-incorporated micelles in the hydrogel-bearing mouse model. These findings indicate that fibrin hydrogels have a strong promoting effect on the recruitment of anti-inflammatory macrophages. Therefore, fibrin hydrogels may be an optimal biomaterial in the design of medicines for macrophage-induced regenerative therapies. STATEMENT OF SIGNIFICANCE: The immune response to tissue injury is important for determining the speed and the result of the regeneration. Alternatively activated macrophages (M2 macrophages) resolve inflammatory response and promote tissue repair by producing anti-inflammatory factors. Promoting the recruitment of macrophages is a hopeful strategy in the design of biomaterials for tissue regeneration. In the present study, we combined the fibrin hydrogel, which promotes anti-inflammatory polarization, with a macrophage recruitment agent. We revealed that the fibrin hydrogel significantly promoted anti-inflammatory polarization in mouse in vivo and human in vitro. Moreover, macrophages significantly infiltrated into the fibrin hydrogel regardless of the agent combination. Fibrin hydrogels may become a reliable biomaterial for tissue regeneration, and the present study is believed to provide information for many researchers.
Collapse
|
188
|
Zhu Y, Jiang P, Luo B, Lan F, He J, Wu Y. Dynamic protein corona influences immune-modulating osteogenesis in magnetic nanoparticle (MNP)-infiltrated bone regeneration scaffolds in vivo. NANOSCALE 2019; 11:6817-6827. [PMID: 30912535 DOI: 10.1039/c8nr08614a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
An inflammatory reaction initiates fracture healing and directly influences the osteoinductive effect of the magnetic hydroxyapatite (MHA) scaffold, but the underlying mechanism is yet to be elucidated. Protein corona as a real biological identity of a biomaterial significantly affects the biological function of the bone regenerative scaffold. Hence, we developed a simple and effective in vivo dynamic model for the protein corona of MHA scaffolds to predict the correlation between the inflammatory reaction and bone wound healing, as well as the underlying mechanism governing such a process. Certain proteins including proteins related to the immune response and inflammation, bone and wound healing, extracellular matrix, cell behavior, and signaling increased in the protein corona of the magnetic nanoparticle (MNP)-infiltrated scaffolds in a time-dependent manner. Moreover, the enriched proteins related to the immune response and inflammation adsorbed on the MHA scaffolds correlated well with the proteins that significantly enhanced bone wound healing, as suggested by the same variation tendency of the proteins related to bone and wound healing, and immune response and inflammation. The presence of MNPs suppressed the chronic inflammatory responses and highly promoted the acute inflammatory responses. More importantly, the activation of the acute inflammatory responses led to the recruitment of immune cells, remodeling of the extracellular matrix and even the acceleration of bone healing. The bone repair in vivo model and inflammatory cytokine in vitro model results further corroborated the critical involvement of inflammatory reaction in enhancing bone wound healing. This opens up the great potential of protein corona formation to understand the complicated mechanisms involved in immune-modulated bone wound healing.
Collapse
Affiliation(s)
- Yue Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, P.R. China.
| | | | | | | | | | | |
Collapse
|
189
|
Macrophage in vitro Response on Hybrid Coatings Obtained by Matrix Assisted Pulsed Laser Evaporation. COATINGS 2019. [DOI: 10.3390/coatings9040236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The improvement in the research area of the implant by surface functionalization when correlated with the biological response is of major interest in the biomedical field. Based on the fact that the inflammatory response is directly involved in the ultimate response of the implant within the body, it is essential to study the macrophage-material interactions. Within this context, we have investigated the composite material-macrophage cell interactions and the inflammatory response to these composites with amorphous hydroxyapatite (HA), Lactoferrin (Lf), and polyethylene glycol-polycaprolactone (PEG-PCL) copolymer. All materials are obtained by Matrix Assisted Pulsed Laser Evaporation (MAPLE) technique and characterized by Atomic Force Microscopy and Scanning Electron Microscopy. Macrophage-differentiated THP-1 cells proliferation and metabolic activity were assessed by qualitative and quantitative methods. The secretion of tumor necrosis factor alpha (TNF-α) and interleukin 10 (IL-10) cytokine, in the presence and absence of the inflammatory stimuli (bacterial endotoxin; lipopolysaccharide (LPS)), was measured using an ELISA assay. Our results revealed that the cellular response depended on the physical-chemical characteristics of the coatings. Copolymer-HA-Lf coatings led to low level of pro-inflammatory TNF-α, the increased level of anti-inflammatory IL-10, and the polarization of THP-1 cells towards an M2 pro-reparative phenotype in the presence of LPS. These findings could have important potential for the development of composite coatings in implant applications.
Collapse
|
190
|
Veiseh O, Vegas AJ. Domesticating the foreign body response: Recent advances and applications. Adv Drug Deliv Rev 2019; 144:148-161. [PMID: 31491445 PMCID: PMC6774350 DOI: 10.1016/j.addr.2019.08.010] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 01/03/2023]
Abstract
The foreign body response is an immunological process that leads to the rejection of implanted devices and presents a fundamental challenge to their performance, durability, and therapeutic utility. Recent advances in materials development and device design are now providing strategies to overcome this immune-mediated reaction. Here, we briefly review our current mechanistic understanding of the foreign body response and highlight new anti-FBR technologies from this decade that have been applied successfully in biomedical applications relevant to implants, devices, and cell-based therapies. Further development of these important technologies promises to enable new therapies, diagnostics, and revolutionize the management of patient care for many intractable diseases.
Collapse
Affiliation(s)
- Omid Veiseh
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77030, USA.
| | - Arturo J Vegas
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA.
| |
Collapse
|
191
|
Comparison of the host macrophage response to synthetic and biologic surgical meshes used for ventral hernia repair. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.regen.2018.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
192
|
Liao Y, Ouyang L, Ci L, Chen B, Lv D, Li Q, Sun Y, Fei J, Bao S, Liu X, Li L. Pravastatin regulates host foreign-body reaction to polyetheretherketone implants via miR-29ab1-mediated SLIT3 upregulation. Biomaterials 2019; 203:12-22. [PMID: 30851489 DOI: 10.1016/j.biomaterials.2019.02.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
Abstract
Host rejection to biomaterials can induce uncontrolled foreign-body reactions (FBR), resulting in a dense fibrous encapsulation that blocks mass transport and/or communication between the host and the implant. Adequate angiogenesis between the body and the implant has been implicated as a key regulator for overcoming FBR. Thus, approaches for stimulating neovascularization and/or suppressing FBR are under investigation. In this study, pravastatin (Pra) was loaded onto a 3D network surface of sulfonated polyetheretherketone (SP) to achieve superior local drug effects. The SP loaded with Pra (SP-Pra) promoted angiogenesis and mitigated FBR via miR-29 dependent SLIT3 upregulation in wild-type (WT) mice. miR-29a and miR-29b1 were significantly downregulated in the SP-Pra capsule compared to levels in the SP capsule, while SLIT3 and neovascularization were substantially upregulated in WT mice. However, the above effects presented in the WT mice were not detected in miR-29ab1 knockout mice which was generated by the CRISPR/Cas9 approach. Overall, the results suggest that miR-29 plays a critical role in reducing FBR to these implants by targeting SLIT3. Suppression of FBR by SP-Pra implants offers the potential to improve the performance of current medical devices.
Collapse
Affiliation(s)
- Yun Liao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Liping Ouyang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai 201203, China
| | - Baohui Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dan Lv
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Qin Li
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yingxiao Sun
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Shisan Bao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; Discipline of Pathology, Charles Perkin Centre, Bosch Institute and School of Medical Sciences, The University of Sydney, Australia.
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Ling Li
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| |
Collapse
|
193
|
Madonna R, Van Laake LW, Botker HE, Davidson SM, De Caterina R, Engel FB, Eschenhagen T, Fernandez-Aviles F, Hausenloy DJ, Hulot JS, Lecour S, Leor J, Menasché P, Pesce M, Perrino C, Prunier F, Van Linthout S, Ytrehus K, Zimmermann WH, Ferdinandy P, Sluijter JPG. ESC Working Group on Cellular Biology of the Heart: position paper for Cardiovascular Research: tissue engineering strategies combined with cell therapies for cardiac repair in ischaemic heart disease and heart failure. Cardiovasc Res 2019; 115:488-500. [PMID: 30657875 PMCID: PMC6383054 DOI: 10.1093/cvr/cvz010] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/15/2022] Open
Abstract
Morbidity and mortality from ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and are increasing worldwide. Patients with IHD or HF might benefit from novel therapeutic strategies, such as cell-based therapies. We recently discussed the therapeutic potential of cell-based therapies and provided recommendations on how to improve the therapeutic translation of these novel strategies for effective cardiac regeneration and repair. Despite major advances in optimizing these strategies with respect to cell source and delivery method, the clinical outcome of cell-based therapy remains unsatisfactory. Major obstacles are the low engraftment and survival rate of transplanted cells in the harmful microenvironment of the host tissue, and the paucity or even lack of endogenous cells with repair capacity. Therefore, new ways of delivering cells and their derivatives are required in order to empower cell-based cardiac repair and regeneration in patients with IHD or HF. Strategies using tissue engineering (TE) combine cells with matrix materials to enhance cell retention or cell delivery in the transplanted area, and have recently received much attention for this purpose. Here, we summarize knowledge on novel approaches emerging from the TE scenario. In particular, we will discuss how combinations of cell/bio-materials (e.g. hydrogels, cell sheets, prefabricated matrices, microspheres, and injectable matrices) combinations might enhance cell retention or cell delivery in the transplantation areas, thereby increase the success rate of cell therapies for IHD and HF. We will not focus on the use of classical engineering approaches, employing fully synthetic materials, because of their unsatisfactory material properties which render them not clinically applicable. The overall aim of this Position Paper from the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to proceed in research with these novel TE strategies combined with cell-based therapies to boost cardiac repair in the clinical settings of IHD and HF.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Institute of Cardiology and Center of Excellence on Aging, “G. d’Annunzio” University—Chieti, Italy
- University of Texas Medical School in Houston, USA
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Raffaele De Caterina
- Institute of Cardiology and Center of Excellence on Aging, “G. d’Annunzio” University—Chieti, Italy
- University of Texas Medical School in Houston, USA
- University of Pisa, Pisa University Hospital, Pisa, Italy
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Muscle Research Center Erlangen, MURCE
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Francesco Fernandez-Aviles
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London, UK
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
| | - Jean-Sebastien Hulot
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
- Paris Cardiovascular Research Center (PARCC), INSERM UMRS 970, Paris, France
- Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Sandrine Lecour
- Hatter Cardiovascular Research Institute, University of Cape Town, South Africa
| | - Jonathan Leor
- Tamman and Neufeld Cardiovascular Research Institutes, Sackler Faculty of Medicine, Tel-Aviv University and Sheba Medical Center, Tel-Hashomer, Israel
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
- INSERM UMRS 970, Paris, France
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Fabrice Prunier
- Institut Mitovasc, INSERM, CNRS, Université d’Angers, Service de Cardiologie, CHU Angers, Angers, France
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany
- Department of Cardiology, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT, The Arctic University of Norway, Norway
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, III-V Floor, H-1089 Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX Utrecht, the Netherlands
| |
Collapse
|
194
|
Huynh V, Jesmer AH, Shoaib MM, Wylie RG. Influence of Hydrophobic Cross-Linkers on Carboxybetaine Copolymer Stimuli Response and Hydrogel Biological Properties. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:1631-1641. [PMID: 30558419 DOI: 10.1021/acs.langmuir.8b03908] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Poly(carboxybetaine) (pCB) hydrogels do not elicit a foreign body response due to their low-fouling properties, making them ideal implantable materials for in vivo drug and cell delivery. Current reported pCB hydrogels are cross-linked using cytotoxic UV-initiated radical polymerization limiting clinical and in vivo translation. For clinical translation, we require in situ and biorthogonal cross-linking of pCB hydrogels that are both low-fouling and low-swelling to limit nonspecific interactions and minimize tissue damage, respectively. To this end, we synthesized carboxybetaine (CB) random copolymers (molecular weight (MW): ∼7-33 kDa; Đ: 1.1-1.36) containing azide (pCB-azide) or strained alkyne (Dibenzocyclooctyne (DBCO); pCB-DBCO) that rapidly cross-link upon mixing. Unlike CB homopolymers and other CB copolymers studied, high DBCO content pCB-DBCO30 (30% DBCO mole fraction) is thermoresponsive with a upper critical solution temperature (UCST; cloud point of ∼20 °C at 50 g/L) in water due to electrostatic associations. Due to the antipolyelectrolyte effect, pCB-DBCO30 is salt-responsive and is soluble even at low temperatures in 5 M NaCl, which prevents zwitterion electrostatic associations. pCB-azide and pCB-DBCO with 0.05 to 0.16 cross-linker mole fractions rapidly formed 10 wt % hydrogels upon mixing that were low-swelling (increase of ∼10% in wet weight) while remaining low-fouling to proteins (∼10-20 μg cm-2) and cells, making them suitable for in vivo applications. pCB-X31 hydrogels composed of pCB-azide32 and pCB-DBCO30 formed opaque gels in water and physiological conditions that shrunk to ∼70% of their original wet weight due to pCB-DBCO30's greater hydrophobicity and interchain electrostatic interactions, which promotes nonspecific protein adsorption (∼35 μg cm-2) and cell binding. Once formed, the electrostatic interactions in pCB-X31 hydrogels are not fully reversible with heat or salt. Although, pCB-X31 hydrogels are transparent when initially prepared in 5 M NaCl. This is the first demonstration of a thermo- and salt-responsive CB copolymer that can tune hydrogel protein and cell fouling properties.
Collapse
Affiliation(s)
- Vincent Huynh
- Department of Chemistry and Chemical Biology , McMaster University , Hamilton , Ontario L8S 4M1 , Canada
| | - Alexander H Jesmer
- Department of Chemistry and Chemical Biology , McMaster University , Hamilton , Ontario L8S 4M1 , Canada
| | - Muhammad M Shoaib
- Department of Chemistry and Chemical Biology , McMaster University , Hamilton , Ontario L8S 4M1 , Canada
| | - Ryan G Wylie
- Department of Chemistry and Chemical Biology , McMaster University , Hamilton , Ontario L8S 4M1 , Canada
| |
Collapse
|
195
|
Oldenkamp HF, Vela Ramirez JE, Peppas NA. Re-evaluating the importance of carbohydrates as regenerative biomaterials. Regen Biomater 2019; 6:1-12. [PMID: 30740237 PMCID: PMC6362819 DOI: 10.1093/rb/rby023] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/20/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Heidi F Oldenkamp
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Julia E Vela Ramirez
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
196
|
Sun W, Luo Z, Lee J, Kim HJ, Lee K, Tebon P, Feng Y, Dokmeci MR, Sengupta S, Khademhosseini A. Organ-on-a-Chip for Cancer and Immune Organs Modeling. Adv Healthc Mater 2019; 8:e1801363. [PMID: 30605261 PMCID: PMC6424124 DOI: 10.1002/adhm.201801363] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/07/2018] [Indexed: 12/21/2022]
Abstract
Bridging the gap between findings in preclinical 2D cell culture models and in vivo tissue cultures has been challenging; the simple microenvironment of 2D monolayer culture systems may not capture the cellular response to drugs accurately. Three-dimensional organotypic models have gained increasing interest due to their ability to recreate precise cellular organizations. These models facilitate investigation of the interactions between different sub-tissue level components through providing physiologically relevant microenvironments for cells in vitro. The incorporation of human-sourced tissues into these models further enables personalized prediction of drug responses. Integration of microfluidic units into the 3D models can be used to control their local environment, dynamic simulation of cell behaviors, and real-time readout of drug testing data. Cancer and immune system related diseases are severe burdens to our health care system and have created an urgent need for high-throughput, and effective drug development plans. This review focuses on recent progress in the development of "cancer-on-a-chip" and "immune organs-on-a-chip" systems designed to study disease progression and predict drug-induced responses. Future challenges and opportunities are also discussed.
Collapse
Affiliation(s)
- Wujin Sun
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Zhimin Luo
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Junmin Lee
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Han-Jun Kim
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - KangJu Lee
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Peyton Tebon
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA
| | - Yudi Feng
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; College of Chemistry, Nankai University, Tianjin 300071, China
| | - Mehmet R. Dokmeci
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Shiladitya Sengupta
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, ; Harvard – MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA, ; Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California-Los Angleles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California - Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90024, USA.; Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA; Department of Radiology, University of California-Los Angeles, Los Angeles, CA 90095, USA; Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia; Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
197
|
Biomaterials: Foreign Bodies or Tuners for the Immune Response? Int J Mol Sci 2019; 20:ijms20030636. [PMID: 30717232 PMCID: PMC6386828 DOI: 10.3390/ijms20030636] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
The perspectives of regenerative medicine are still severely hampered by the host response to biomaterial implantation, despite the robustness of technologies that hold the promise to recover the functionality of damaged organs and tissues. In this scenario, the cellular and molecular events that decide on implant success and tissue regeneration are played at the interface between the foreign body and the host inflammation, determined by innate and adaptive immune responses. To avoid adverse events, rather than the use of inert scaffolds, current state of the art points to the use of immunomodulatory biomaterials and their knowledge-based use to reduce neutrophil activation, and optimize M1 to M2 macrophage polarization, Th1 to Th2 lymphocyte switch, and Treg induction. Despite the fact that the field is still evolving and much remains to be accomplished, recent research breakthroughs have provided a broader insight on the correct choice of biomaterial physicochemical modifications to tune the reaction of the host immune system to implanted biomaterial and to favor integration and healing.
Collapse
|
198
|
Rahmati M, Mozafari M. Biological Response to Carbon-Family Nanomaterials: Interactions at the Nano-Bio Interface. Front Bioeng Biotechnol 2019; 7:4. [PMID: 30729107 PMCID: PMC6351449 DOI: 10.3389/fbioe.2019.00004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/04/2019] [Indexed: 02/06/2023] Open
Abstract
During the last few decades, several studies have suggested that carbon-based nanomaterials, owing to their unique properties, could act as promising candidates in biomedical engineering application. Wide-ranging research efforts have investigated the cellular and molecular responses to carbon-based nanomaterials at the nano-bio interfaces. In addition, a number of surface functionalization strategies have been introduced to improve their safety profile in the biological environment. The present review discusses the general principles of immunological responses to nanomaterials. Then, it explains essential physico-chemical properties of carbon-familynanomaterials, including carbon nanotubes (CNTs), graphene, fullerene, carbon quantum dots (CDs), diamond-like carbon (DLC), and mesoporous carbon biomaterials (MCNs), which significantly affect the immunological cellular and molecular responses at the nano-bio interface. The discussions also briefly highlight the recent studies that critically investigated the cellular and molecular responses to various carbon-based nanomaterials. It is expected that the most recent perspective strategies for improving the biological responses to carbon-based nanomaterials can revolutionize their functions in emerging biological applications.
Collapse
Affiliation(s)
- Maryam Rahmati
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
199
|
Shang W, Chen G, Li Y, Zhuo Y, Wang Y, Fang Z, Yu Y, Ren H. Static Magnetic Field Accelerates Diabetic Wound Healing by Facilitating Resolution of Inflammation. J Diabetes Res 2019; 2019:5641271. [PMID: 31886281 PMCID: PMC6915019 DOI: 10.1155/2019/5641271] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/22/2019] [Indexed: 12/15/2022] Open
Abstract
Impaired wound healing is commonly encountered in patients with diabetes mellitus, which may lead to severe outcomes such as amputation, if untreated timely. Macrophage plays a critical role in the healing process including the resolution phase. Although magnetic therapy is known to improve microcirculation, its effect on wound healing remains uncertain. In the present study, we found that 0.6 T static magnetic field (SMF) significantly accelerated wound closure and elevated reepithelialization and revascularization in diabetic mice. Notably, SMF promoted the wound healing by skewing the macrophage polarization towards M2 phenotype, thus facilitating the resolution of inflammation. In addition, SMF upregulated anti-inflammatory gene expression via activating STAT6 and suppressing STAT1 in macrophage. Taken together, our results indicate that SMF may be a promising adjuvant therapeutic tool for treating diabetic wounds.
Collapse
Affiliation(s)
- Wenlong Shang
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Guilin Chen
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yinxiu Li
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yujuan Zhuo
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuhong Wang
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhicai Fang
- Heye Health Industrial Research Institute of Zhejiang Heye Health Technology, Anji, Zhejiang 313300, China
| | - Ying Yu
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huiwen Ren
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
200
|
Li YM, Wu JY, Jiang J, Dong SK, Chen YS, He HY, Liu CS, Zhao JZ. Chondroitin sulfate-polydopamine modified polyethylene terephthalate with extracellular matrix-mimetic immunoregulatory functions for osseointegration. J Mater Chem B 2019; 7:7756-7770. [DOI: 10.1039/c9tb01984g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Optimal integration between the polyethylene terephthalate (PET) graft and host bone is a prerequisite to obtain a satisfactory outcome after graft implantation for ligament reconstruction.
Collapse
Affiliation(s)
- Ya-Min Li
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Jing-Yao Wu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Jia Jiang
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Shi-Kui Dong
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Yun-Su Chen
- Department of Joint Surgery
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Hong-Yan He
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Chang-Sheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Jin-Zhong Zhao
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| |
Collapse
|