1
|
Sánchez-López L, Chico B, García-Alonso MC, Lozano RM. Macrophage proteomic analysis of covalent immobilization of hyaluronic acid and graphene oxide on CoCr alloy in a tribocorrosive environment. J Biomed Mater Res A 2024; 112:1941-1959. [PMID: 38775427 DOI: 10.1002/jbm.a.37751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 09/03/2024]
Abstract
In this work, a sequential covalent immobilization of graphene oxide (GO) and hyaluronic acid (HA) is performed to obtain a biocompatible wear-resistant nanocoating on the surface of the biomedical grade cobalt-chrome (CoCr) alloy. Nanocoated CoCr surfaces were characterized by Raman spectroscopy and electrochemical impedance spectroscopy (EIS) in 3 g/L HA electrolyte. Tribocorrosion tests of the nanocoated CoCr surfaces were carried out in a pin on flat tribometer. The biological response of covalently HA/GO biofunctionalized CoCr surfaces with and without wear-corrosion processes was studied through the analysis of the proteome of macrophages. Raman spectra revealed characteristic bands of GO and HA on the functionalized CoCr surfaces. The electrochemical response by EIS showed a stable and protective behavior over 23 days in the simulated biological environment. HA/GO covalently immobilized on CoCr alloy is able to protect the surface and reduce the wear volume released under tribocorrosion tests. Unsupervised classification analysis of the macrophage proteome via hierarchical clustering and principal component analysis (PCA) revealed that the covalent functionalization on CoCr enhances the macrophage biocompatibility in vitro. On the other hand, disruption of the HA/GO nanocoating by tribocorrosion processes induced a macrophage proteome which was differently clustered and was distantly located in the PCA space. In addition, tribocorrosion induced an increase in the percentage of upregulated and downregulated proteins in the macrophage proteome, revealing that disruption of the covalent nanocoating impacts the macrophage proteome. Although macrophage inflammation induced by tribocorrosion of HA/GO/CoCr surfaces is observed, it is ameliorated by the covalently grafting of HA, which provides immunomodulation by eliciting downregulations in characteristic pro-inflammatory signaling involved in inflammation and aseptic loosening of CoCr joint arthroplasties. Covalent HA/GO nanocoating on CoCr provides potential applications for in vivo joint prostheses led a reduced metal-induced inflammation and degradation by wear-corrosion.
Collapse
Affiliation(s)
- L Sánchez-López
- Centro de Investigaciones Biológicas-Margarita Salas (CIB Margarita Salas), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro Nacional de Investigaciones Metalúrgicas (CENIM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- PhD Program in Advanced Materials and Nanotechnology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain
| | - B Chico
- Centro Nacional de Investigaciones Metalúrgicas (CENIM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Maria Cristina García-Alonso
- Centro Nacional de Investigaciones Metalúrgicas (CENIM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Rosa M Lozano
- Centro de Investigaciones Biológicas-Margarita Salas (CIB Margarita Salas), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
2
|
Wang S, Chen Y, Ling Z, Li J, Hu J, He F, Chen Q. The role of dendritic cells in the immunomodulation to implanted biomaterials. Int J Oral Sci 2022; 14:52. [PMCID: PMC9636170 DOI: 10.1038/s41368-022-00203-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Considering the substantial role played by dendritic cells (DCs) in the immune system to bridge innate and adaptive immunity, studies on DC-mediated immunity toward biomaterials principally center on their adjuvant effects in facilitating the adaptive immunity of codelivered antigens. However, the effect of the intrinsic properties of biomaterials on dendritic cells has not been clarified. Recently, researchers have begun to investigate and found that biomaterials that are nonadjuvant could also regulate the immune function of DCs and thus affect subsequent tissue regeneration. In the case of proteins adsorbed onto biomaterial surfaces, their intrinsic properties can direct their orientation and conformation, forming “biomaterial-associated molecular patterns (BAMPs)”. Thus, in this review, we focused on the intrinsic physiochemical properties of biomaterials in the absence of antigens that affect DC immune function and summarized the underlying signaling pathways. Moreover, we preliminarily clarified the specific composition of BAMPs and the interplay between some key molecules and DCs, such as heat shock proteins (HSPs) and high mobility group box 1 (HMGB1). This review provides a new direction for future biomaterial design, through which modulation of host immune responses is applicable to tissue engineering and immunotherapy.
Collapse
Affiliation(s)
- Siyuan Wang
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Yanqi Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Zhaoting Ling
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jia Li
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jun Hu
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Fuming He
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Qianming Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| |
Collapse
|
3
|
Yang Y, Lin Y, Xu R, Zhang Z, Zeng W, Xu Q, Deng F. Micro/Nanostructured Topography on Titanium Orchestrates Dendritic Cell Adhesion and Activation via β2 Integrin-FAK Signals. Int J Nanomedicine 2022; 17:5117-5136. [PMID: 36345509 PMCID: PMC9636866 DOI: 10.2147/ijn.s381222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
Background and Purpose In clinical application of dental implants, the functional state of dendritic cells (DCs) has been suggested to have a close relationship with the implant survival rate or speed of osseointegration. Although microscale surfaces have a stable osteogenesis property, they also incline to trigger unfavorable DCs activation and threaten the osseointegration process. Nanoscale structures have an advantage in regulating cell immune response through orchestrating cell adhesion, indicating the potential of hierarchical micro/nanostructured surface in regulation of DCs’ activation without sacrificing the advantage of microscale topography. Materials and Methods Two micro/nanostructures were fabricated based on microscale rough surfaces through anodization or alkali treatment, the sand-blasted and acid-etched (SA) surface served as control. The surface characteristics, in vitro and in vivo DC immune reactions and β2 integrin-FAK signal expression were systematically investigated. The DC responses to different surface topographies after FAK inhibition were also tested. Results Both micro/nano-modified surfaces exhibited unique composite structures, with higher hydrophilicity and lower roughness compared to the SA surface. The DCs showed relatively immature functional states with round morphologies and significantly downregulated β2 integrin-FAK levels on micro/nanostructures. Implant surfaces with micro/nano-topographies also triggered lower levels of DC inflammatory responses than SA surfaces in vivo. The inhibited FAK activation effectively reduced the differences in topography-caused DC activation and narrowed the differences in DC activation among the three groups. Conclusion Compared to the SA surface with solely micro-scale topography, titanium surfaces with hybrid micro/nano-topographies reduced DC inflammatory response by influencing their adhesion states. This regulatory effect was accompanied by the modulation of β2 integrin-FAK signal expression. The β2 integrin-FAK-mediated adhesion plays a critical role in topography-induced DC activation, which represents a potential target for material–cell interaction regulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yujing Lin
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Ruogu Xu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Zhengchuan Zhang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Wenyi Zeng
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Qiong Xu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China,Correspondence: Qiong Xu; Feilong Deng, Department of Oral Implantology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, No. 56, Ling Yuan Xi Road, Guangzhou, 510055, People’s Republic of China, Tel +86 20 83862537, Fax +86 20 83822807, Email ;
| | - Feilong Deng
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| |
Collapse
|
4
|
Eslami-Kaliji F, Mirahmadi-Zare SZ, Nazem S, Shafie N, Ghaedi R, Asadian-Esfahani MH. A label-free SPR biosensor for specific detection of TLR4 expression; introducing of 10-HDA as an antagonist. Int J Biol Macromol 2022; 217:142-149. [PMID: 35817233 DOI: 10.1016/j.ijbiomac.2022.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 11/05/2022]
Abstract
Toll-like receptor 4 (TLR4) is actively involved in many health-related problems, including transplantation rejection and autoimmune diseases. Therefore, it is important to identify an antagonist to inhibit the TLR4-induced immune cell activation. In our previous study, 10-hydroxy-2-decanoic acid (10-HDA) was introduced as a potential antagonist for TLR4; however, possible interaction between 10-HDA and TLR4 needed to be detected. Due to the ability of surface plasmon resonance (SPR) biosensor to confirm the specific interactions between receptors and ligands, a new configuration of SPR biosensor proposed to detect the possible interaction between 10-HDA and TLR4. Hence, 10-HDA was immobilized using the (3-aminopropyl) triethoxysilane (APTES) polymer as a crosslinking agent on the Ag-MgF2 surface. Besides, genetically modified HEK293T cells with high TLR4 expression were used to study the possible interaction between 10-HDA and TLR4. Surprisingly, the SPR angle was significantly reduced in the presence of HEK cells expressing TLR4, while HEK cells without TLR4 did not affect the SPR angle. So, the proposed SPR biosensor successfully detected the interaction betweenTLR4 and 10-HDA. The sensitivity and detection limit of the biosensor were achieved at 0.05 and 0.5 million cells expressing TLR4, respectively, with a two-fold dynamic range.
Collapse
Affiliation(s)
- Farshid Eslami-Kaliji
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Seyede Zohreh Mirahmadi-Zare
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran.
| | - Saeid Nazem
- Department of Physics, Faculty of Science, University of Isfahan, Isfahan, Iran
| | - Negar Shafie
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Rassoul Ghaedi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | | |
Collapse
|
5
|
Abudula T, Colombani T, Alade T, Bencherif SA, Memić A. Injectable Lignin- co-Gelatin Cryogels with Antioxidant and Antibacterial Properties for Biomedical Applications. Biomacromolecules 2021; 22:4110-4121. [PMID: 34514795 DOI: 10.1021/acs.biomac.1c00575] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
For several biomedical applications, it is essential to develop novel bioactive materials. Such biomaterials could potentially improve wound healing, prevent infections, or be used in immunoengineering. For example, bioactive materials that reduce oxidative stress without relying on antibiotics and other drugs could be beneficial. Hydrogel-based biomaterials, especially those derived from natural polymers, have been regarded as one of the most promising scaffolds for biomedical research. These multifunctional scaffolds can exhibit high water adsorption capacity, biocompatibility, and biomechanical properties that can match native tissues. Cryogels are a special type of hydrogels in which polymers are cross-linked around ice crystals. As a result, cryogels exhibit unique physical features, including a macroporous and interconnected network, flexibility, shape-memory properties, and syringe injectability. Herein, we developed a multifunctional, i.e., antibacterial, antioxidant, and injectable cryogel by combining lignin with gelatin. The cryogel with 0.2% lignin showed a compressive modulus of 25 kPa and a compressive stress of 140 kPa at 80% strain, which is, respectively, 1.8 and 7 times higher than those of the pure gelatin cryogels. Meanwhile, such a cryogel formulation could completely recover its shape after compression up to 90% and was needle-injectable. Additionally, the lignin-co-gelatin cryogel with 0.1-0.2 lignin showed 8-10 mm of inhibition zone against the most common surgical site infection-associated pathogenic bacteria. Furthermore, lignin-co-gelatin cryogel was found to scavenge free radicals and have good cytocompatibility, and the cryogels with up to 0.2% lignin minimally activate naïve mouse bone marrow-derived dendritic cells. Overall, the current approach shows great promise for the design of bioresource-based multifunctional cryogels for a wide range of biomedical applications.
Collapse
Affiliation(s)
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Taofeek Alade
- Center of Nanotechnology, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Sidi A Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States.,Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States.,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States.,UMR CNRS 7338 Biomechanics and Bioengineering, University of Technology of Compiègne, Sorbonne University, 60200 Compiègne, France
| | - Adnan Memić
- Center of Nanotechnology, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
6
|
Abaricia JO, Farzad N, Heath TJ, Simmons J, Morandini L, Olivares-Navarrete R. Control of innate immune response by biomaterial surface topography, energy, and stiffness. Acta Biomater 2021; 133:58-73. [PMID: 33882355 DOI: 10.1016/j.actbio.2021.04.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
As the focus of implantable biomaterials has shifted from bioinert implants to bioactive designs, recent research has highlighted the complex interactions between cell physiologic systems and material properties, particularly physical cues. From the cells known to interact with implanted biomaterials, the response of the immune system has been a critical target of study recently. Here, we review studies characterizing the response of innate immune cells to various material cues, particularly of those at the surface of implanted materials.The innate immune system consists of cell types with various roles in inflammation. Neutrophils and macrophages serve both phagocytic and signaling roles, especially early in the inflammatory phase of biomaterial implantation. These cell types ultimately dictate the outcome of implants as chronic inflammation, fibrosis, or integration. Other cell types like dendritic cells, mast cells, natural killer cells, and innate lymphoid cells may also serve an immunomodulatory role in the biomaterial context. This review highlights recent advances in our understanding of the role of innate immunity in the response to implantable biomaterials as well as key mechanobiological findings in innate immune cells underpinning these advances. STATEMENT OF SIGNIFICANCE: This review highlights recent advances in the understanding of the role of innate immunity in the response to implantable biomaterials, especially in neutrophils and macrophages, as well as key mechanobiological findings in innate immune cells underpinning these advances. Here we discuss how physicochemical properties of biomaterials control innate immune cell behavior.
Collapse
|
7
|
Eslami-Kaliji F, Sarafbidabad M, Kiani-Esfahani A, Mirahmadi-Zare SZ, Dormiani K. 10-hydroxy-2-decenoic acid a bio-immunomodulator in tissue engineering; generates tolerogenic dendritic cells by blocking the toll-like receptor4. J Biomed Mater Res A 2021; 109:1575-1587. [PMID: 33638611 DOI: 10.1002/jbm.a.37152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs), in response to the biomaterials, utilize toll-like receptors (TLRs) to become mature or tolerogenic through TLRs-dependent signaling pathways, especially TLR4. Regarding the physicochemical properties of biomaterials, some of such signaling pathways are activated. Unsaturated fatty acids have been explored as an antagonist for TLRs and lead to the tolerogenic phenotype of DCs. Here we showed that, although cultured DCs on both chitosan and Alginate-polyethyleneimine (Alg-PEI) films became fully mature, 10-hydroxy-2-decanoic acid (10-HDA), an unsaturated fatty acid found in royal jelly, led to the tolerogenic immunophenotype of DCs on both films. The cultured cells on the films possessed iDCs-like morphology in the presence of 10-HDA. Moreover, 10-HDA expressed lower levels of CD80, CD83, CD86, and HLA-DR, a higher level of IL-10, and lower level of IL-12 in the cultured DCs on both films. Furthermore, HEK293T cells expressing only TLR4 (HEK-TLR4 cells) were co-cultured with LPS, a specific agonist for TLR4, and 10-HDA. The 10-HDA significantly reduced the expression of tumor necrosis factor-a (TNF-α) in the HEK-TLR4 cells compared to treated only with LPS. These findings indicate that the 10-HDA acts as an antagonist of TLR4; therefore, potentially can be used in autoimmune diseases and preventing the rejection of biomaterials implantation and allograft transplantation.
Collapse
Affiliation(s)
- Farshid Eslami-Kaliji
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran.,Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Sarafbidabad
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Abbas Kiani-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Seyede Zohreh Mirahmadi-Zare
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kianoush Dormiani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
8
|
Batool F, Özçelik H, Stutz C, Gegout PY, Benkirane-Jessel N, Petit C, Huck O. Modulation of immune-inflammatory responses through surface modifications of biomaterials to promote bone healing and regeneration. J Tissue Eng 2021; 12:20417314211041428. [PMID: 34721831 PMCID: PMC8554547 DOI: 10.1177/20417314211041428] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/06/2021] [Indexed: 12/25/2022] Open
Abstract
Control of inflammation is indispensable for optimal oral wound healing and tissue regeneration. Several biomaterials have been used to enhance the regenerative outcomes; however, the biomaterial implantation can ensure an immune-inflammatory response. The interface between the cells and the biomaterial surface plays a critical role in determining the success of soft and hard tissue regeneration. The initial inflammatory response upon biomaterial implantation helps in tissue repair and regeneration, however, persistant inflammation impairs the wound healing response. The cells interact with the biomaterials through extracellular matrix proteins leading to protein adsorption followed by recruitment, attachment, migration, and proliferation of several immune-inflammatory cells. Physical nanotopography of biomaterials, such as surface proteins, roughness, and porosity, is crucial for driving cellular attachment and migration. Similarly, modification of scaffold surface chemistry by adapting hydrophilicity, surface charge, surface coatings, can down-regulate the initiation of pro-inflammatory cascades. Besides, functionalization of scaffold surfaces with active biological molecules can down-regulate pro-inflammatory and pro-resorptive mediators' release as well as actively up-regulate anti-inflammatory markers. This review encompasses various strategies for the optimization of physical, chemical, and biological properties of biomaterial and the underlying mechanisms to modulate the immune-inflammatory response, thereby, promoting the tissue integration and subsequent soft and hard tissue regeneration potential of the administered biomaterial.
Collapse
Affiliation(s)
- Fareeha Batool
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Hayriye Özçelik
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Céline Stutz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Pierre-Yves Gegout
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Pôle de médecine et chirurgie bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Catherine Petit
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Pôle de médecine et chirurgie bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Huck
- Faculté de Chirurgie-dentaire, Université de Strasbourg, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Pôle de médecine et chirurgie bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
9
|
Srinivasan S, Babensee JE. Controlled Delivery of Immunomodulators from a Biomaterial Scaffold Niche to Induce a Tolerogenic Phenotype in Human Dendritic Cells. ACS Biomater Sci Eng 2020; 6:4062-4076. [DOI: 10.1021/acsbiomaterials.0c00439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sangeetha Srinivasan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Julia E. Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
10
|
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
|
11
|
Beigi MH, Safaie N, Nasr-Esfahani MH, Kiani A. 3D Titania Nanofiber-Like Webs Induced by Plasma Ionization: A New Direction for Bioreactivity and Osteoinductivity Enhancement of Biomaterials. Sci Rep 2019; 9:17999. [PMID: 31784696 PMCID: PMC6884481 DOI: 10.1038/s41598-019-54533-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022] Open
Abstract
In this study, we describe the formation method of web-like three-dimensional (3-D) titania nanofibrous structures coated on transparent substrate via a high intensity laser induced reverse transfer (HILIRT) process. First, we demonstrate the mechanism of ablation and deposition of Ti on the glass substrates using multiple picosecond laser pulses at ambient air in an explicit analytical form and compare the theoretical results with the experimental results of generated nanofibers. We then examine the performance of the developed glass samples coated by titania nanofibrous structures at varied laser pulse durations by electron microscopy and characterization methods. We follow this by exploring the response of human bone-derived mesenchymal stem cells (BMSCs) with the specimens, using a wide range of in-vitro analyses including MTS assay (colorimetric method for assessing cell metabolic activity), immunocytochemistry, mineralization, ion release examination, gene expression analysis, and protein adsorption and absorption analysis. Our results from the quantitative and qualitative analyses show a significant biocompatibility improvement in the laser treated samples compared to untreated substrates. By decreasing the pulse duration, more titania nanofibers with denser structures can be generated during the HILIRT technique. The findings also suggest that the density of nanostructures and concentration of coated nanofibers play critical roles in the bioreactivity properties of the treated samples, which results in early osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Mohammad-Hossein Beigi
- Silicon Hall: Micro/Nano Manufacturing Facility, Faculty of Engineering and Applied Science, Ontario Tech University, Ontario, Canada
- Department of Cellular Biotechnology Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Naghmeh Safaie
- Silicon Hall: Micro/Nano Manufacturing Facility, Faculty of Engineering and Applied Science, Ontario Tech University, Ontario, Canada
| | - Mohammad-Hossein Nasr-Esfahani
- Department of Cellular Biotechnology Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Amirkianoosh Kiani
- Silicon Hall: Micro/Nano Manufacturing Facility, Faculty of Engineering and Applied Science, Ontario Tech University, Ontario, Canada.
| |
Collapse
|
12
|
Zhu FJ, Tong YL, Sheng ZY, Yao YM. Role of dendritic cells in the host response to biomaterials and their signaling pathways. Acta Biomater 2019; 94:132-144. [PMID: 31108257 DOI: 10.1016/j.actbio.2019.05.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022]
Abstract
Strategies to enhance, inhibit, or qualitatively modulate immune responses are important for diverse biomedical applications such as vaccine adjuvant, drug delivery, immunotherapy, cell transplant, tissue engineering, and regenerative medicine. However, the clinical efficiency of these biomaterial systems is affected by the limited understanding of their interaction with complex host microenvironments, for example, excessive foreign body reaction and immunotoxicity. Biomaterials and biomedical devices implanted in the body may induce a highly complicated and orchestrated series of host responses. As macrophages are among the first cells to infiltrate and respond to implanted biomaterials, the macrophage-mediated host response to biomaterials has been well studied. Dendritic cells (DCs) are the most potent antigen-presenting cells that activate naive T cells and bridge innate and adaptive immunity. The potential interaction of DCs with biomaterials appears to be critical for exerting the function of biomaterials and has become an important, developing area of investigation. Herein, we summarize the effects of the physicochemical properties of biomaterials on the immune function of DCs together with their receptors and signaling pathways. This review might provide a complete understanding of the interaction of DCs with biomaterials and serve as a reference for the design and selection of biomaterials with particular effects on targeted cells. STATEMENT OF SIGNIFICANCE: Biomaterials implanted in the body are increasingly applied in clinical practice. The performance of these implanted biomaterials is largely dependent on their interaction with the host immune system. As antigen-presenting cells, dendritic cells (DCs) directly interact with biomaterials through pattern recognition receptors (PRRs) recognizing "biomaterial-associated molecular patterns" and generate a battery of immune responses. In this review, the physicochemical properties of biomaterials that regulate the immune function of DCs together with their receptors and signaling pathways of biomaterial-DC interactions are summarized and discussed. We believe that knowledge of the interplay of DC and biomaterials may spur clinical translation by guiding the design and selection of biomaterials with particular effects on targeted cell for tissue engineering, vaccine delivery, and cancer therapy.
Collapse
|
13
|
Macrophage response to hydrophilic biomaterials regulates MSC recruitment and T-helper cell populations. Biomaterials 2018; 182:202-215. [PMID: 30138783 DOI: 10.1016/j.biomaterials.2018.08.029] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 08/04/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022]
Abstract
Successful biomaterial implantation can be achieved by controlling the activation of the immune system. The innate immune system is typically the focus on synthetic material compatibility, but this study shows an effect of surface properties in the innate as well as the adaptive systems. These studies look at how macrophages respond to the implanted materials by releasing factors to regulate the microenvironment and recruit additional cells. Our research demonstrates how macrophage response to material surface properties can create changes in the adaptive immune response by altering T-helper cell populations and stem cell recruitment. Titanium (Ti) implants of varying wettability (rough, and rough-hydrophilic) were placed in the femur of 10-week-old male C57Bl/6, or macrophage ablated clodronate liposome injected and transgenic MaFIA (C57BL/6-Tg(Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6)2Bck/J) mice. The microenvironment surrounding Ti implants was assessed using custom PCR arrays at 3 and 7 days following implantation. Changes in specific T-helper, macrophage and stem cell populations were evaluated locally at the implant surface and systemically in the contralateral leg bone marrow and spleen by flow cytometry at 1, 3 and 7 days. Macrophage importance in T-helper and stem cell population changes with metallic surfaces was examined in both in vitro and in vivo with macrophage ablation models. We demonstrate that surface modifications applied to titanium implants to increase surface roughness and wettability can polarize the adaptive immune response towards a Th2, pro-wound healing phenotype, leading to faster resolution of inflammation and increased stem cell recruitment around rough hydrophilic implants with macrophages present.
Collapse
|
14
|
Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S, Formenti SC. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer 2018; 18:313-322. [PMID: 29449659 PMCID: PMC5912991 DOI: 10.1038/nrc.2018.6] [Citation(s) in RCA: 764] [Impact Index Per Article: 127.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
More than 60 years ago, the effect whereby radiotherapy at one site may lead to regression of metastatic cancer at distant sites that are not irradiated was described and called the abscopal effect (from 'ab scopus', that is, away from the target). The abscopal effect has been connected to mechanisms involving the immune system. However, the effect is rare because at the time of treatment, established immune-tolerance mechanisms may hamper the development of sufficiently robust abscopal responses. Today, the growing consensus is that combining radiotherapy with immunotherapy provides an opportunity to boost abscopal response rates, extending the use of radiotherapy to treatment of both local and metastatic disease. In this Opinion article, we review evidence for this growing consensus and highlight emerging limitations to boosting the abscopal effect using immunotherapy. This is followed by a perspective on current and potential cross-disciplinary approaches, including the use of smart materials to address these limitations.
Collapse
Affiliation(s)
- Wilfred Ngwa
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, 450 Brookline Avenue, Boston, MA, USA
| | - Omoruyi Credit Irabor
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, 450 Brookline Avenue, Boston, MA, USA
| | - Jonathan D. Schoenfeld
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, 450 Brookline Avenue, Boston, MA, USA
| | - Jürgen Hesser
- University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3. D-68167, Mannheim, Germany
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, 1300 York Avenue, Box 169, New York, NY, USA
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, 1300 York Avenue, Box 169, New York, NY, USA
| |
Collapse
|
15
|
Paschoalin RT, Traldi B, Aydin G, Oliveira JE, Rütten S, Mattoso LH, Zenke M, Sechi A. Solution blow spinning fibres: New immunologically inert substrates for the analysis of cell adhesion and motility. Acta Biomater 2017; 51:161-174. [PMID: 28069500 DOI: 10.1016/j.actbio.2017.01.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 12/31/2022]
Abstract
The control of cell behaviour through material geometry is appealing as it avoids the requirement for complex chemical surface modifications. Significant advances in new technologies have been made to the development of polymeric biomaterials with controlled geometry and physico-chemical properties. Solution blow spinning technique has the advantage of ease of use allowing the production of nano or microfibres and the direct fibre deposition on any surface in situ. Yet, in spite of these advantages, very little is known about the influence of such fibres on biological functions such as immune response and cell migration. In this work, we engineered polymeric fibres composed of either pure poly(lactic acid) (PLA) or blends of PLA and polyethylene glycol (PEG) by solution blow spinning and determined their impact on dendritic cells, highly specialised cells essential for immunity and tolerance. We also determined the influence of fibres on cell adhesion and motility. Cells readily interacted with fibres resulting in an intimate contact characterised by accumulation of actin filaments and focal adhesion components at sites of cell-fibre interactions. Moreover, cells were guided along the fibres and actin and focal adhesion components showed a highly dynamic behaviour at cell-fibre interface. Remarkably, fibres did not elicit any substantial increase of activation markers and inflammatory cytokines in dendritic cells, which remained in their immature (inactive) state. Taken together, these findings will be useful for developing new biomaterials for applications in tissue engineering and regenerative medicine.
Collapse
|
16
|
Leifer CA. Dendritic cells in host response to biologic scaffolds. Semin Immunol 2017; 29:41-48. [PMID: 28214177 DOI: 10.1016/j.smim.2017.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/23/2017] [Accepted: 01/31/2017] [Indexed: 02/03/2023]
Abstract
Tissue regeneration and repair require a highly complex and orchestrated series of events that require inflammation, but can be compromised when inflammation is excessive or becomes chronic. Macrophages are one of the first cells to contact and respond to implanted materials, and mediate the inflammatory response. The series of events following macrophage association with biomaterials has been well-studied. Dendritic cells (DCs) also directly interact with biomaterials, are critical for specific immune responses, and can be activated in response to interactions with biomaterials. Yet, much less is known about the responses by DCs. This review discusses what we know about DC response to biomaterials, the underlying mechanisms involved, and how DCs can be influenced by the macrophage response to biomaterials. Lastly, I will discuss how biomaterials can be manipulated to enhance or suppress DC function to promote a specific desirable immune response - a major goal for implantable biologically active therapeutics.
Collapse
Affiliation(s)
- Cynthia A Leifer
- Department of Microbiology and Immunology College of Veterinary Medicine, C5-153 Cornell University, Ithaca, NY, USA.
| |
Collapse
|
17
|
Keselowsky BG, Lewis JS. Dendritic cells in the host response to implanted materials. Semin Immunol 2017; 29:33-40. [PMID: 28487131 PMCID: PMC5612375 DOI: 10.1016/j.smim.2017.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/29/2017] [Accepted: 04/18/2017] [Indexed: 12/28/2022]
Abstract
The role of dendritic cells (DCs) and their targeted manipulation in the body's response to implanted materials is an important and developing area of investigation, and a large component of the emerging field of biomaterials-based immune engineering. The key position of DCs in the immune system, serving to bridge innate and adaptive immunity, is facilitated by rich diversity in type and function and places DCs as a critical mediator to biomaterials of both synthetic and natural origins. This review presents current views regarding DC biology and summarizes recent findings in DC responses to implanted biomaterials. Based on these findings, there is promise that the directed programming of application-specific DC responses to biomaterials can become a reality, enabling and enhancing applications almost as diverse as the larger field of biomaterials itself.
Collapse
Affiliation(s)
- Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611 USA.
| | - Jamal S Lewis
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| |
Collapse
|
18
|
Xu P, Tang S, Jiang L, Yang L, Zhang D, Feng S, Zhao T, Dong Y, He W, Wang R, Zhang J, Liang Z. Nanomaterial-dependent immunoregulation of dendritic cells and its effects on biological activities of contraceptive nanovaccines. J Control Release 2016; 225:252-68. [PMID: 26826303 DOI: 10.1016/j.jconrel.2016.01.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 01/19/2016] [Accepted: 01/24/2016] [Indexed: 01/08/2023]
Abstract
Nanovehicles are promising delivery systems for various vaccines. Nevertheless, different biophysicochemical properties of nanoparticles (NPs), dominating their in vitro and in vivo performances for vaccination, remain unclear. We attempted to elucidate the effects of NPs and their pH-sensitivity on in vitro and in vivo efficacy of resulting prophylactic nanovaccines containing a contraceptive peptide (FSHR). To this end, pH-responsive and non-responsive nanovaccines were produced using acetalated β-cyclodextrin (Ac-bCD) and poly(lactic-co-glycolic acid) (PLGA), respectively. Meanwhile, FSHR derived from an epitope of the follicle-stimulating hormone receptor was used as the model antigen. FSHR-containing Ac-bCD and PLGA NPs were successfully prepared by a nanoemulsion technique, leading to well-shaped nanovaccines with high loading efficiency. The pH-sensitivity of Ac-bCD and PLGA nanovaccines was examined by in vitro hydrolysis and antigen release studies. Nanovaccines could be effectively engulfed by dendritic cells (DCs) via endocytosis in both dose and time dependent manners, and their intracellular trafficking was closely related to the pH-sensitivity of the carrier materials. Furthermore, nanovaccines could induce the secretion of inflammatory cytokines by DCs and T cells co-cultured with the stimulated DCs. In vivo evaluations demonstrated that nanovaccines were more potent than that based on the complete Freund's adjuvant, with respect to inducing anti-FSHR antibody, reducing the sperm count, inhibiting the sperm motility, and increasing the teratosperm rate. Immunization of male mice with nanovaccines notably decreased the parturition incidence of the mated females. Consequently, both in vitro and in vivo activities of FSHR could be considerably augmented by NPs. More importantly, our studies indicated that the pH-responsive nanovaccine was not superior over the non-responsive counterpart for the examined peptide antigen.
Collapse
Affiliation(s)
- Pingping Xu
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; Department of Obstetrics and Gynaecology, Hospital 81 of PLA, Nanjing 210002, China
| | - Shuai Tang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Luping Jiang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lihua Yang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; Department of Obstetrics and Gynaecology, Tangshan Workers' Hospital of Hebei Province, Tangshan 063000, China
| | - Dinglin Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Shibin Feng
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Tingting Zhao
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Yajun Dong
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei He
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| | - Zhiqing Liang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
19
|
Abstract
Strategies to enhance, suppress, or qualitatively shape the immune response are of importance for diverse biomedical applications, such as the development of new vaccines, treatments for autoimmune diseases and allergies, strategies for regenerative medicine, and immunotherapies for cancer. However, the intricate cellular and molecular signals regulating the immune system are major hurdles to predictably manipulating the immune response and developing safe and effective therapies. To meet this challenge, biomaterials are being developed that control how, where, and when immune cells are stimulated in vivo, and that can finely control their differentiation in vitro. We review recent advances in the field of biomaterials for immunomodulation, focusing particularly on designing biomaterials to provide controlled immunostimulation, targeting drugs and vaccines to lymphoid organs, and serving as scaffolds to organize immune cells and emulate lymphoid tissues. These ongoing efforts highlight the many ways in which biomaterials can be brought to bear to engineer the immune system.
Collapse
Affiliation(s)
- Nathan A Hotaling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
| | - Li Tang
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
| | - Darrell J Irvine
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| | - Julia E Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia 30332;
| |
Collapse
|
20
|
Sumida A, Gogas BD, Nagai H, Li J, King SB, Chronos N, Hou D. A comparison of drug eluting stent biocompatibility between third generation NOBORI biolimus A9-eluting stent and second generation XIENCE V everolimus-eluting stent in a porcine coronary artery model. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2015; 16:351-7. [DOI: 10.1016/j.carrev.2015.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 06/06/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
|
21
|
Zhu H, Yang F, Tang B, Li XM, Chu YN, Liu YL, Wang SG, Wu DC, Zhang Y. Mesenchymal stem cells attenuated PLGA-induced inflammatory responses by inhibiting host DC maturation and function. Biomaterials 2015; 53:688-98. [DOI: 10.1016/j.biomaterials.2015.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/29/2022]
|
22
|
Sauter A, Mc Duffie Y, Boehm H, Martinez A, Spatz JP, Appel S. Surface-mediated priming during in vitro generation of monocyte-derived dendritic cells. Scand J Immunol 2015; 81:56-65. [PMID: 25376441 DOI: 10.1111/sji.12246] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/16/2014] [Accepted: 10/21/2014] [Indexed: 11/28/2022]
Abstract
Ex vivo-generated human dendritic cells (DC) are most commonly generated from monocytes using standard cell culture dishes. To elucidate the effect of the plastic surface during the differentiation process, we compared a standard adhesive plastic dish with four different mainly non-adherent surfaces. Untouched monocytes were cultured for 3 days in the presence of IL-4 and GM-CSF. Time-lapse videos were recorded, and the phenotype of the cells was analysed by flow cytometry. The cytokine profiles were analysed using a 25-plex cytokine assay. The use of non-adherent surfaces led to a significant reduction in expression of CD14 and CD38, and a significant increase in expression of CD86 compared to standard culture dishes. Expression levels of DC-SIGN and PD-L2 were reduced significantly on cells cultured on non-adherent surfaces. The cytokine production was independent on the surface used. The surface-mediated priming should therefore be considered when aiming to induce specific immune responses. This is especially important with regard to DC-based immunotherapy, where an adjustment of the surface during the DC generation process might have highly beneficial effects.
Collapse
Affiliation(s)
- A Sauter
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | | | | | | | |
Collapse
|
23
|
Park J, Gerber MH, Babensee JE. Phenotype and polarization of autologous T cells by biomaterial-treated dendritic cells. J Biomed Mater Res A 2014; 103:170-84. [PMID: 24616366 DOI: 10.1002/jbm.a.35150] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/06/2014] [Accepted: 02/19/2014] [Indexed: 12/27/2022]
Abstract
Given the central role of dendritic cells (DCs) in directing T-cell phenotypes, the ability of biomaterial-treated DCs to dictate autologous T-cell phenotype was investigated. In this study, we demonstrate that differentially biomaterial-treated DCs differentially directed autologous T-cell phenotype and polarization, depending on the biomaterial used to pretreat the DCs. Immature DCs (iDCs) were derived from human peripheral blood monocytes and treated with biomaterial films of alginate, agarose, chitosan, hyaluronic acid, or 75:25 poly(lactic-co-glycolic acid) (PLGA), followed by co-culture of these biomaterial-treated DCs and autologous T cells. When autologous T cells were co-cultured with DCs treated with biomaterial film/antigen (ovalbumin, OVA) combinations, different biomaterial films induced differential levels of T-cell marker (CD4, CD8, CD25, CD69) expression, as well as differential cytokine profiles [interferon (IFN)-γ, interleukin (IL)-12p70, IL-10, IL-4] in the polarization of T helper (Th) types. Dendritic cells treated with agarose films/OVA induced CD4+CD25+FoxP3+ (T regulatory cells) expression, comparable to untreated iDCs, on autologous T cells in the DC-T co-culture system. Furthermore, in this co-culture, agarose treatment induced release of IL-12p70 and IL-10 at higher levels as compared with DC treatment with other biomaterial films/OVA, suggesting Th1 and Th2 polarization, respectively. Dendritic cells treated with PLGA film/OVA treatment induced release of IFN-γ at higher levels compared with that observed for co-cultures with iDCs or DCs treated with all other biomaterial films. These results indicate that DC treatment with different biomaterial films has potential as a tool for immunomodulation by directing autologous T-cell responses.
Collapse
Affiliation(s)
- Jaehyung Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia, 30332
| | | | | |
Collapse
|
24
|
Lewis JS, Dolgova N, Chancellor T, Acharya AP, Karpiak JV, Lele TP, Keselowsky BG. The effect of cyclic mechanical strain on activation of dendritic cells cultured on adhesive substrates. Biomaterials 2013; 34:9063-70. [PMID: 24008042 PMCID: PMC4120880 DOI: 10.1016/j.biomaterials.2013.08.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/09/2013] [Indexed: 12/15/2022]
Abstract
Dendritic cells (DCs), key regulators of tolerance and immunity, have been found to reside in mechanically active tissues such as the interior layers of the arterial wall, which experience cyclic radial wall strain due to pulsatile blood flow. Although experimentally difficult to determine in vivo, it is reasonable to postulate DCs experience the mechanical forces in such mechanically active tissues. However, it is currently unknown how DCs respond to cyclic mechanical strain. In order to explore the hypothesis that DCs are responsive to mechanical strain, DCs were cultured in vitro on pre-adsorbed adhesive proteins (e.g., laminin, collagen, fibrinogen) and 1 Hz cyclic strain was applied for various durations and strain magnitudes. It was determined that a strain magnitude of 10% and 24 h duration adversely affected DC viability compared to no-strain controls, but culture on certain adhesive substrates provided modest protection of viability under this harsh strain regime. In contrast, application of 1 h of 1 Hz cyclic 3% strain did not affect DC viability and this strain regime was used for the remaining experiments for quantifying DC activation and T-cell priming capability. Application of 3% strain increased expression of stimulatory (MHC-II) and costimulatory molecules (CD86, CD40), and this effect was generally increased by culture on pre-coated adhesive substrates. Interestingly, the cytokine secretion profile of DCs was not significantly affected by strain. Lastly, strained DCs demonstrated increased stimulation of allogeneic T-cell proliferation, in a manner that was independent of the adhesive substrate. These observations indicate generation of a DC consistent with what has been described as a semi-mature phenotype. This work begins elucidating a potential role for DCs in tissue environments exposed to cyclic mechanical forces.
Collapse
Affiliation(s)
- Jamal S. Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611 U.S
| | - Natalia Dolgova
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611 U.S
| | - T.J. Chancellor
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611 U.S
| | - Abhinav P. Acharya
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611 U.S
| | - Jerome V. Karpiak
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611 U.S
| | - Tanmay P. Lele
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611 U.S
| | - Benjamin G. Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611 U.S
| |
Collapse
|
25
|
Bayrak A, Prüger P, Stock UA, Seifert M. Absence of immune responses with xenogeneic collagen and elastin. Tissue Eng Part A 2013; 19:1592-600. [PMID: 23406399 DOI: 10.1089/ten.tea.2012.0394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Novel tissue-engineering approaches for cardiovascular matrices based on xenogeneic extracellular matrix protein (ECMp) constituents require a detailed evaluation of their interaction with essential immune cell subsets playing a role in innate or adaptive immunity. Therefore, in this study, the effects of xenogeneic (porcine, bovine) collagen type I and elastin as the two main components of the heart valve ECM were analyzed in comparison to their human equivalents. First, their potential to induce maturation and cytokine secretion of human dendritic cells (DC) was tested by flow cytometry. Second, the influence on proliferation and cytokine release of purified human B and T cells was measured. We could demonstrate that xenogeneic collagen type I and elastin are not able to trigger the maturation of DC as verified by the lack of CD83 induction accompanied by a low tumor necrosis factor-α release. Moreover, both ECMp showed no effect on the proliferation and the interleukin-6 release of either unstimulated or prestimulated B cells. Additionally, anti-CD3-induced purified T cell proliferation and secretion of cytokines was not affected. All in vitro data verify the low immunogenicity of porcine and bovine collagen type I and elastin and favor their suitability for tissue-engineered scaffolds.
Collapse
Affiliation(s)
- Alexandra Bayrak
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
26
|
Park J, Babensee JE. Differential functional effects of biomaterials on dendritic cell maturation. Acta Biomater 2012; 8:3606-17. [PMID: 22705044 DOI: 10.1016/j.actbio.2012.06.006] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 06/01/2012] [Accepted: 06/06/2012] [Indexed: 12/18/2022]
Abstract
The immunological outcome of dendritic cell (DC) treatment with different biomaterials was assessed to demonstrate the range of DC phenotypes induced by biomaterials commonly used in combination products. Immature DCs (iDCs) were derived from human peripheral blood monocytes, and treated with different biomaterial films of alginate, agarose, chitosan, hyaluronic acid (HA), or 75:25 poly(lactic-co-glycolic acid) (PLGA) and a comprehensive battery of phenotypic functional outcomes was assessed. Different levels of functional changes in DC phenotype were observed depending on the type of biomaterial films used to treat the DCs. Treatment of DCs with PLGA or chitosan films supported DC maturation, with higher levels of DC allostimulatory capacity, pro-inflammatory cytokine release, and expression of CD80, CD86, CD83, HLA-DQ and CD44 compared with iDCs, and lower endocytic ability compared with iDCs. Alginate film induced pro-inflammatory cytokine release from DCs at levels higher than from iDCs. Dendritic cells treated with HA film expressed lower levels of CD40, CD80, CD86 and HLA-DR compared with iDCs. They also exhibited lower endocytic ability and CD44 expression than iDCs, possibly due to an insolubilized (cross-linked) form of high molecular weight HA. Interestingly, treatment of DCs with agarose film maintained the DC functional phenotype at levels similar to iDCs except for CD44 expression, which was lower than that of iDCs. Taken together, these results can provide selection criteria for biomaterials to be used in immunomodulating applications and can inform potential outcomes of biomaterials within combination products on associated immune responses as desired by the application.
Collapse
|
27
|
Pathak SK, Sköld AE, Mohanram V, Persson C, Johansson U, Spetz AL. Activated apoptotic cells induce dendritic cell maturation via engagement of Toll-like receptor 4 (TLR4), dendritic cell-specific intercellular adhesion molecule 3 (ICAM-3)-grabbing nonintegrin (DC-SIGN), and β2 integrins. J Biol Chem 2012; 287:13731-42. [PMID: 22396536 DOI: 10.1074/jbc.m111.336545] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells playing a central role in connecting innate and adaptive immunity. Maturation signals are, however, required for DCs to undergo phenotypic and functional changes to acquire a fully competent antigen-presenting capacity. We previously reported that activated apoptotic peripheral lymphocytes (ActApo) provide activation/maturation signals to human monocyte-derived DCs. In this paper, we have characterized the signaling pathways and molecules involved in ActApo-mediated DC maturation. We found that both cellular and supernatant fractions from ActApo are required for DC maturation signaling. ActApoSup-induced CD80 and CD86 expression was significantly blocked in the presence of neutralizing antibodies against tumor necrosis factor-α (TNF-α). Cell-cell contact-dependent signaling involved β2 integrins, dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN), and TLR4 because ActApo-induced up-regulation of the maturation markers CD80 and CD86 was significantly inhibited in the presence of neutralizing antibodies against CD18, CD11a, CD11b, and DC-SIGN as well as TLR4. The role of TLR4 was further confirmed by silencing of TLR4 in DCs. In addition, the endogenous adjuvant effect exerted by activated apoptotic splenocytes (ActApoSp) was reduced after immunization with human serum albumin in TLR4(-/-) mice. We detected activation of multiple signaling pathways and transcription factors in DCs upon co-culture with ActApo, including p38, JNK, PI3K-Akt, Src family kinases, NFκB p65, and AP1 transcription factor family members c-Jun and c-Fos, demonstrating the complex interactions occurring between ActApo and DCs. These studies provide important mechanistic insight into the responses of DCs during encounter with cells undergoing immunogenic cell death.
Collapse
Affiliation(s)
- Sushil Kumar Pathak
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, S-141 86 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
28
|
Hume PS, He J, Haskins K, Anseth KS. Strategies to reduce dendritic cell activation through functional biomaterial design. Biomaterials 2012; 33:3615-25. [PMID: 22361099 DOI: 10.1016/j.biomaterials.2012.02.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 02/02/2012] [Indexed: 10/28/2022]
Abstract
Dendritic cells play a key role in determining adaptive immunity, and there is growing interest in characterizing and manipulating the interactions between dendritic cells and biomaterial surfaces. Contact with several common biomaterials can induce the maturation of immature dendritic cells, but substrates that reduce dendritic cell maturation are of particular interest within the field of cell-based therapeutics where the goal is to reduce the immune response to cell-laden material carriers. In this study, we use a materials-based strategy to functionalize poly(ethylene glycol) hydrogels with immobilized immunosuppressive factors (TGF-β1 and IL-10) to reduce the maturation of immature dendritic cells. TGF-β1 and IL-10 are commonly employed as soluble factors to program dendritic cells in vitro, and we demonstrate that these proteins retain bioactivity towards dendritic cells when immobilized on hydrogel surfaces. Following stimulation with lipopolysaccharide (LPS) and/or cytokines, a dendritic cell line interacting with the surfaces of immunosuppressive hydrogels expressed reduced markers of maturation, including IL-12 and MHCII. The bioactivity of these immunomodulatory hydrogels was further confirmed with primary bone marrow-derived dendritic cells (BMDCs) isolated from non-obese diabetic (NOD) mice, as quantified by a decrease in activation markers and a significantly reduced capacity to activate T cells. Furthermore, by introducing a second signal to promote BMDC-material interactions combined with the presentation of tolerizing signals, the multifunctional PEG hydrogels were found to further increase signaling towards BMDCs, as evidenced by greater reductions in maturation markers.
Collapse
Affiliation(s)
- Patrick S Hume
- Department of Chemical and Biological Engineering, University of Colorado, 424 UCB, Boulder, CO 80309, USA
| | | | | | | |
Collapse
|
29
|
van den Dries K, van Helden SFG, te Riet J, Diez-Ahedo R, Manzo C, Oud MM, van Leeuwen FN, Brock R, Garcia-Parajo MF, Cambi A, Figdor CG. Geometry sensing by dendritic cells dictates spatial organization and PGE(2)-induced dissolution of podosomes. Cell Mol Life Sci 2011; 69:1889-901. [PMID: 22204022 PMCID: PMC3350765 DOI: 10.1007/s00018-011-0908-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 11/28/2011] [Accepted: 12/13/2011] [Indexed: 01/01/2023]
Abstract
Assembly and disassembly of adhesion structures such as focal adhesions (FAs) and podosomes regulate cell adhesion and differentiation. On antigen-presenting dendritic cells (DCs), acquisition of a migratory and immunostimulatory phenotype depends on podosome dissolution by prostaglandin E2 (PGE2). Whereas the effects of physico-chemical and topographical cues have been extensively studied on FAs, little is known about how podosomes respond to these signals. Here, we show that, unlike for FAs, podosome formation is not controlled by substrate physico-chemical properties. We demonstrate that cell adhesion is the only prerequisite for podosome formation and that substrate availability dictates podosome density. Interestingly, we show that DCs sense 3-dimensional (3-D) geometry by aligning podosomes along the edges of 3-D micropatterned surfaces. Finally, whereas on a 2-dimensional (2-D) surface PGE2 causes a rapid increase in activated RhoA levels leading to fast podosome dissolution, 3-D geometric cues prevent PGE2-mediated RhoA activation resulting in impaired podosome dissolution even after prolonged stimulation. Our findings indicate that 2-D and 3-D geometric cues control the spatial organization of podosomes. More importantly, our studies demonstrate the importance of substrate dimensionality in regulating podosome dissolution and suggest that substrate dimensionality plays an important role in controlling DC activation, a key process in initiating immune responses.
Collapse
Affiliation(s)
- Koen van den Dries
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cornelius RM, Shankar SP, Brash JL, Babensee JE. Immunoblot analysis of proteins associated with self-assembled monolayer surfaces of defined chemistries. J Biomed Mater Res A 2011; 98:7-18. [PMID: 21509932 PMCID: PMC3155773 DOI: 10.1002/jbm.a.33084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 11/02/2010] [Accepted: 01/07/2011] [Indexed: 11/06/2022]
Abstract
Intact and fragmented proteins, eluted from self-assembled monolayer (SAM) surfaces of alkanethiols of different chemistries (-CH₃, -OH, -COOH, -NH₂), following exposure to human plasma (HP) or human serum (HS), were examined using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting techniques. The SAM surfaces were incubated for 1 h with 10% (v/v) sterile-filtered, heat-inactivated (h.i.) HS or 1% (v/v) sterile-filtered h.i. HP preparations [both in phosphate buffered saline (PBS)]. Adsorbed proteins were eluted using 10% SDS/2.3% dithioerythritol for characterization of protein profiles. The type of incubating medium may be an important determinant of adsorbed protein profiles, since some variations were observed in eluates from filtered versus control unfiltered h.i. 10% HS or 1% HP. Albumin and apolipoprotein A1 were consistently detected in both filtered h.i 10% HS and 1% HP eluates from all SAM surfaces and from control tissue culture-treated polystyrene (TCPS). Interestingly, Factor H and Factor I, antithrombin, prothrombin, high molecular weight kininogen (HMWK), and IgG were present in eluates from OH, COOH, and NH₂ SAM surfaces and in eluates from TCPS but not in eluates from CH₃ SAM surfaces, following exposure to filtered h.i. 10% HS. These results suggest that CH₃ SAM surfaces were the least proinflammatory of all SAM surfaces. Overall, similar trends were observed in the profiles of proteins eluted from surfaces exposed to filtered 10% HS or 1% HP. However, the unique profiles of adsorbed proteins on different SAM surface chemistries may be related to their differential interactions with cells, including immune/inflammatory cells.
Collapse
Affiliation(s)
- Rena M. Cornelius
- Department of Chemical Engineering and School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Sucharita P. Shankar
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University 313 Ferst Drive Atlanta, GA 30332 USA
| | - John L. Brash
- Department of Chemical Engineering and School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Julia E. Babensee
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University 313 Ferst Drive Atlanta, GA 30332 USA
| |
Collapse
|