Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Biomed Mater Res A ; 112(8): 1317-1336, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38562052

ABSTRACT

Translational research in biomaterials and immunoengineering is leading to the development of novel advanced therapeutics to treat diseases such as cancer, autoimmunity, and viral infections. Dendritic cells (DCs) are at the center of these therapeutics given that they bridge innate and adaptive immunity. The biomaterial system developed herein uses a hydrogel carrier to deliver immunomodulatory DCs for amelioration of autoimmunity. This biomaterial vehicle is comprised of a poly (ethylene glycol)-4 arm maleimide (PEG-4MAL) hydrogels, conjugated with the immunosuppressive cytokine, interleukin-10, IL-10, and cross-linked with a collagenase-degradable peptide sequence for the injectable delivery of immunosuppressive DCs to an anatomical disease-relevant site of the cervical lymph nodes, for intended application to treat multiple sclerosis. The amount of IL-10 incorporated in the hydrogel was optimized to be 500 ng in vitro, based on immunological endpoints. At this concentration, DCs exhibited the best viability, most immunosuppressive phenotype, and protection against proinflammatory insult as compared with hydrogel-incorporated DCs with lower IL-10 loading amounts. Additionally, the effect of the degradability of the PEG-4MAL hydrogel on the release rate of incorporated IL-10 was assessed by varying the ratio of degradable peptides: VPM (degradable) and DTT (nondegradable) and measuring the IL-10 release rates. This IL-10-conjugated hydrogel delivery system for immunosuppressive DCs is set to be assessed for in vivo functionality as the immunosuppressive cytokine provides a tolerogenic environment that keeps DCs in their immature phenotype, which consequently enhances cell viability and optimizes the system's immunomodulatory functionality.


Subject(s)
Dendritic Cells , Hydrogels , Interleukin-10 , Polyethylene Glycols , Polyethylene Glycols/chemistry , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Hydrogels/chemistry , Interleukin-10/metabolism , Animals , Mice , Mice, Inbred C57BL , Humans
2.
ACS Biomater Sci Eng ; 8(10): 4341-4353, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36134725

ABSTRACT

Biomaterial systems such as hydrogels enable localized delivery and postinjection modulation of cellular therapies in a wide array of contexts. Biomaterials as adjuvants have been an active area of investigation, but the study of functionalized biomaterials supporting immunosuppressive cell therapies for tolerogenic applications is still nascent. Here, we developed a 4-arm poly(ethylene-glycol)-maleimide (PEG-4MAL) hydrogel functionalized with interleukin-10 (IL-10) to improve the local delivery and efficacy of a cell therapy against autoimmune disease. The biophysical and biochemical properties of PEG-4MAL hydrogels were optimized to support dendritic cell (DC) viability and an immature phenotype. IL-10-functionalized PEG-4MAL (PEG-IL10) hydrogels exhibited controlled IL-10 release, extended the duration of DC support, and protected DCs from inflammatory assault. After incorporation in PEG-IL10 hydrogels, these DCs induced CD25+FoxP3+ regulatory T cells (Tregs) during in vitro coculture. These studies serve as a proof-of-concept for improving the efficacy of immunosuppressive cell therapies through biomaterial delivery. The flexible nature of this system enables its widespread application across a breadth of other tolerogenic applications for future investigation.


Subject(s)
Hydrogels , Interleukin-10 , Biocompatible Materials/pharmacology , Dendritic Cells/metabolism , Ethylenes , Forkhead Transcription Factors/metabolism , Hydrogels/chemistry , Hydrogels/pharmacology , Maleimides/chemistry , Phenotype , Polyethylene Glycols/chemistry
3.
J Biomed Mater Res A ; 109(7): 1247-1255, 2021 07.
Article in English | MEDLINE | ID: mdl-33040412

ABSTRACT

In multiple sclerosis (MS), abnormally activated immune cells responsive to myelin proteins result in widespread damage throughout the central nervous system (CNS) and ultimately irreversible disability. Immunomodulation by delivering dendritic cells (DCs) utilizes a potent and rapid MS disease progression driver therapeutically. Here, we investigated delivering DCs for disease severity attenuation using an experimental autoimmune encephalomyelitis preclinical MS model. DCs treated with interleukin-10 (IL-10) (DC10s) were transplanted using in situ gelling poly(ethylene glycol)-based hydrogel for target site localization. DC delivery increased hydrogel longevity and altered the injection site recruited, endogenous immune cell profile within 2 days postinjection. Furthermore, hydrogel-mediated DC transplantation efficacy depended on the injection-site. DCs delivered to the neck local to MS-associated CNS-draining cervical lymph nodes attenuated paralysis, compared to untreated controls, while delivery to the flank did not alter paralysis severity. This study demonstrates that local delivery of DC10s modulates immune cell recruitment and attenuates disease progression in a preclinical model of MS.


Subject(s)
Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/therapy , Hydrogels/chemistry , Multiple Sclerosis/therapy , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Polyethylene Glycols/chemistry
4.
ACS Biomater Sci Eng ; 6(7): 4062-4076, 2020 07 13.
Article in English | MEDLINE | ID: mdl-33463332

ABSTRACT

Engineering a regulatory phenotype in dendritic cells (DCs) is a potential approach to circumvent an immune response against self-antigens in autoimmunity or alloantigens in allograft rejection. Cell microenvironments influence the differentiation of DC precursors into either proinflammatory/immunostimulatory or tolerogenic/regulatory DCs. Biomaterial-based vehicles can be used to re-engineer cell microenvironments and re-educate the DC phenotype. This study presents the development and validation of a biomaterial-based multicomponent immunomodulatory (MI) scaffold for the purpose of promoting a tolerogenic/regulatory DC phenotype. Glutaraldehyde-crosslinked gelatin microparticles, loaded with specific immunomodulators, were embedded into a porous agarose scaffold. Using the Weibull equation and the Bayesian approach, an empirical mathematical model was derived from the release profile data of "model" molecules. The scaffold design was generated from the model to achieve distinct temporal release profiles of the loaded immunomodulator(s): granulocyte monocyte colony-stimulating factor (GM-CSF), dexamethasone (DEX), and/or peptidoglycan (PGN). The MI scaffold-treated DCs (MI DCs) showed an increase in the expression of tolerogenic markers such as surface immunoglobulin-like transcript 3 (ILT-3) and secreted interleukin-10 (IL-10), with a simultaneous decrease in maturation markers such as CD86 and secreted interferon-γ (IFN-γ). In cell culture studies, these MI DCs were able to suppress T-cell proliferation. This approach is expected to enhance the generation of endogenous regulatory DCs when applied in vivo. This technology serves as a basis for future immunotherapeutic applications in the autoimmunity and allogeneic therapies. It also shows that empirical mathematical modeling can be used to engineer scaffold designs for distinct temporal release of one or more immunomodulators.


Subject(s)
Biocompatible Materials , Dendritic Cells , Bayes Theorem , Biocompatible Materials/pharmacology , Cells, Cultured , Humans , Phenotype
5.
J Cell Physiol ; 235(6): 5120-5129, 2020 06.
Article in English | MEDLINE | ID: mdl-31674663

ABSTRACT

Dendritic cells are key players in regulating immunity. These cells both activate and inhibit the immune response depending on their cellular environment. Their response to hyperglycemia, a condition common amongst diabetics wherein glucose is abnormally elevated, remains to be elucidated. In this study, the phenotype and immune response of dendritic cells exposed to hyperglycemia were characterized in vitro and in vivo using the streptozotocin-induced diabetes model. Dendritic cells were shown to be sensitive to hyperglycemia both during and after differentiation from bone marrow precursor cells. Dendritic cell behavior under hyperglycemic conditions was found to vary by phenotype, among which, tolerogenic dendritic cells were particularly sensitive. Expression of the costimulatory molecule CD86 was found to reliably increase when dendritic cells were exposed to hyperglycemia. Additionally, hydrogel-based delivery of the anti-inflammatory molecule interleukin-10 was shown to partially inhibit these effects in vivo.


Subject(s)
Dendritic Cells/metabolism , Hyperglycemia/metabolism , Immune Tolerance/genetics , T-Lymphocytes, Regulatory/immunology , Animals , B7-2 Antigen/genetics , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/immunology , Dendritic Cells/pathology , Glucose/metabolism , Humans , Hyperglycemia/genetics , Hyperglycemia/immunology , Hyperglycemia/pathology , Immune Tolerance/immunology , Interleukin-10/pharmacology , Mice , T-Lymphocytes, Regulatory/pathology
6.
Annu Rev Biomed Eng ; 17: 317-49, 2015.
Article in English | MEDLINE | ID: mdl-26421896

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.


Subject(s)
Biocompatible Materials , Immunomodulation , Animals , Antigen-Presenting Cells/immunology , Artificial Cells/immunology , Biomedical Engineering , Dendritic Cells/immunology , Humans , Immunization , Immunologic Factors/administration & dosage , Immunotherapy , Lymphoid Tissue/immunology , Nanoparticles , Nanotechnology , Proteins/immunology , Systems Biology , Tissue Engineering
7.
J Biomed Mater Res A ; 103(1): 170-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24616366

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.


Subject(s)
Biocompatible Materials , Dendritic Cells/cytology , Immunophenotyping , T-Lymphocytes/cytology , Biomarkers/metabolism , Cell Polarity , Coculture Techniques , Dendritic Cells/immunology , Humans
8.
Biomaterials ; 35(22): 5862-74, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24746228

ABSTRACT

Carbohydrates and glycoconjugates have been shown to exert pro-inflammatory effects on the dendritic cells (DCs), supporting pathogen-induced innate immunity and antigen processing, as well as immunosuppressive effects in the tolerance to self-proteins. Additionally, the innate inflammatory response to implanted biomaterials has been hypothesized to be mediated by inflammatory cells interacting with adsorbed proteins, many of which are glycosylated. However, the molecular factors relevant for surface displayed glycoconjugate modulation of dendritic cell (DC) phenotype are unknown. Thus, in this study, a model system was developed to establish the role of glycan composition, density, and carrier cationization state on DC response. Thiol modified glycans were covalently bound to a model protein carrier, maleimide functionalized bovine serum albumin (BSA), and the number of glycans per BSA modulated. Additionally, the carrier isoelectric point was scaled from a pI of ∼4.0 to ∼10.0 using ethylenediamine (EDA). The DC response to the neoglycoconjugates adsorbed to wells of a 384-well plate was determined via a high throughput assay. The underlying trends in DC phenotype in relation to conjugate properties were elucidated via multivariate general linear models. It was found that glycoconjugates with more than 20 glycans per carrier had the greatest impact on the pro-inflammatory response from DCs, followed by conjugates having an isoelectric point above 9.5. Surfaces displaying terminal α1-2 linked mannose structures were able to increase the inflammatory DC response to a greater extent than did any other terminal glycan structure. The results herein can be applied to inform the design of the next generation of combination products and biomaterials for use in future vaccines and implanted materials.


Subject(s)
Dendritic Cells/immunology , Glycoconjugates/chemistry , Glycoconjugates/immunology , Adsorption , Cell Survival , Cells, Cultured , Dendritic Cells/cytology , Humans , Immunity, Innate
9.
Acta Biomater ; 8(10): 3606-17, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705044

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.


Subject(s)
Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Chitosan/pharmacology , Cytokines/metabolism , Dendritic Cells/metabolism , Endocytosis/drug effects , Flow Cytometry , Fluorescence , Histocompatibility Antigens Class II/metabolism , Humans , Hyaluronic Acid/pharmacology , Inflammation Mediators/metabolism , Lactic Acid/pharmacology , Lipopolysaccharides/pharmacology , Photoelectron Spectroscopy , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Receptors, Cell Surface/metabolism , Sepharose/pharmacology , Surface Properties/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Transcription Factor RelA/metabolism
10.
Biomaterials ; 33(6): 1699-713, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22136715

ABSTRACT

Dendritic cells (DCs) play a critical role in orchestrating the host responses to a wide variety of foreign antigens and are essential in maintaining immune tolerance. Distinct biomaterials have been shown to differentially affect the phenotype of DCs, which suggested that biomaterials may be used to modulate immune response toward the biologic component in combination products. The elucidation of biomaterial property-DC phenotype relationships is expected to inform rational design of immuno-modulatory biomaterials. In this study, DC response to a set of 12 polymethacrylates (pMAs) was assessed in terms of surface marker expression and cytokine profile. Principal component analysis (PCA) determined that surface carbon correlated with enhanced DC maturation, while surface oxygen was associated with an immature DC phenotype. Partial square linear regression, a multivariate modeling approach, was implemented and successfully predicted biomaterial-induced DC phenotype in terms of surface marker expression from biomaterial properties with R(prediction)(2) = 0.76. Furthermore, prediction of DC phenotype was effective based on only theoretical chemical composition of the bulk polymers with R(prediction)(2) = 0.80. These results demonstrated that immune cell response can be predicted from biomaterial properties, and computational models will expedite future biomaterial design and selection.


Subject(s)
Biocompatible Materials/chemistry , Coated Materials, Biocompatible/chemistry , Dendritic Cells/cytology , Polymethacrylic Acids/chemistry , Combinatorial Chemistry Techniques , Humans , Kinetics , Materials Testing , Models, Chemical , Multivariate Analysis , Phenotype , Polymers/chemistry , Principal Component Analysis , Regression Analysis , Surface Properties , Temperature , Water/chemistry
11.
J Biomed Mater Res A ; 98(1): 7-18, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21509932

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.


Subject(s)
Blood Proteins/analysis , Immunoblotting/methods , Adsorption , Electrophoresis, Polyacrylamide Gel , Fibrinogen/analysis , Filtration , Humans , Molecular Weight , Polystyrenes , Serum , Surface Properties , Tissue Culture Techniques
12.
Biomaterials ; 32(5): 1270-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21030075

ABSTRACT

Biomaterials have the potential to be utilized as immunostimulatory or immunosuppressive delivery agents for biologics. It is hypothesized that this is directed by the ability of a biomaterial to drive dendritic cells (DC) in situ toward an immunostimulatory or an immunosuppressive phenotype, respectively. However, the specific pattern recognition receptors (PRRs) that DCs use to recognize and respond to biomaterials are unknown. From among the many receptors that DCs use to recognize and respond to foreign entities, herein the focus is on integrins. A biomaterial that induces DC maturation, namely poly(lactic-co-glycolic) acid (PLGA), supported increased human monocyte-derived DC adhesion and up-regulation of integrin receptor gene expression, measured via RT-PCR, as compared to culture on tissue culture polystyrene (TCPS). This was not observed for a biomaterial that does not support DC maturation. Through antibody-blocking techniques, the adhesion to both TCPS and PLGA was found to be ß(2) integrin dependent and ß(1) independent. Significantly, inhibiting ß(2)-mediated adhesion to biomaterials via blocking antibodies also lowered the level of maturation of DCs (CD86 expression). ß(2) integrins (but not ß(1)) were found localized in biomaterial-adherent DC podosomes and also were found in direct contact with the PLGA surface. Therefore, it appeared that ß(2) integrin-mediated adhesion is involved in determining the state of DC maturation on the PLGA surface. DC adhesion to biomaterials may be engaged or avoided to manipulate an immune response to biological component delivered with a biomaterial carrier.


Subject(s)
Biocompatible Materials/pharmacology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Integrins/metabolism , Biocompatible Materials/chemistry , CD18 Antigens/genetics , CD18 Antigens/metabolism , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , Dendritic Cells/drug effects , Fluorescent Antibody Technique , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Lactic Acid/chemistry , Lactic Acid/pharmacology , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Reverse Transcriptase Polymerase Chain Reaction
13.
Acta Biomater ; 7(3): 1354-63, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20977948

ABSTRACT

Dendritic cells (DCs) play pivotal roles in responding to foreign entities during the innate immune response and in initiating effective adaptive immunity as well as maintaining immune tolerance. The sensitivity of DCs to foreign stimuli also makes them useful cells to assess the inflammatory response to biomaterials. Elucidating material property-DC phenotype relationships using a well-defined biomaterial system is expected to provide criteria for immunomodulatory biomaterial design. Clinical titanium (Ti) substrates, including pretreatment (PT), sand blasted and acid etched (SLA), and modified SLA (modSLA), with different roughnesses and surface energies were used to treat DCs and resulted in differential DC responses. PT and SLA induced a mature DC (mDC) phenotype, while modSLA promoted a non-inflammatory environment by supporting an immature DC (iDC) phenotype, based on surface marker expression, cytokine production profiles and cell morphology. Principal component analysis (PCA) confirmed these experimental results, and also indicated that the non-stimulating property of modSLA covaried with certain surface properties, such as high surface hydrophilicity, percent oxygen and percent Ti of the substrates. In addition to previous research that demonstrated superior osteogenic properties of modSLA compared with PT and SLA, the results reported herein indicates that modSLA may further benefit implant osteointegration by reducing local inflammation and its associated osteoclastogenesis.


Subject(s)
Dendritic Cells/immunology , Titanium/chemistry , Cell Differentiation , Cytokines/metabolism , Humans , Microscopy, Electron, Scanning , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Principal Component Analysis , Surface Properties
14.
J Biomed Mater Res A ; 96(1): 239-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21105173

ABSTRACT

Macrophages (Mϕ) and dendritic cells (DCs) are critical antigen presenting cells that play pivotal roles in host responses to biomaterial implants. Although Mϕs have been widely studied for their roles in the inflammatory responses against biomaterials, the roles that DCs play in the host responses toward implanted materials have only recently been explored. DCs are of significant research interest because of the emergence of a large number of combination products that cross-traditional medical device boundaries. These products combine biomaterials with biologics, including cells, nucleic acids, and/or proteins. The biomaterial component may evoke an inflammatory response, primarily mediated by neutrophils and Mϕs, whereas the biologic component may elicit an immunogenic immune response, initiated by DCs involving lymphocyte activation. Control of Mϕ phenotypic balance from proinflammatory M1 to reparative M2 is a goal of investigators to optimize the host response to biomaterials. Similarly, control of DC phenotype from proinflammatory to toleragenic is of interest in vaccine delivery and tissue engineering/transplantation situations, respectively. This review discusses the interconnection between innate and adaptive immunity, the comparative and contrasting phenotypes and roles of Mϕs and DCs in immunity, their responses to biomaterials and the strategies to modulate their phenotype for applications in tissue engineering and vaccine delivery. Furthermore, the collaboration between and unique roles of DCs and Mϕs needs to be addressed in future studies to gain a more complete picture of host responses toward combination products.


Subject(s)
Biocompatible Materials/chemistry , Dendritic Cells/immunology , Macrophages/immunology , Phenotype , Animals , Biocompatible Materials/metabolism , Dendritic Cells/cytology , Immunity/immunology , Macrophage Activation/immunology , Macrophages/cytology , Tissue Engineering
15.
J Control Release ; 146(3): 341-8, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20595029

ABSTRACT

Biomaterials have been shown to differentially support dendritic cell (DC) maturation, a prerequisite for an adjuvant effect. Treatment of DCs with poly(D,L-lactic-co-glycolic acid) (PLGA) films resulted in DC maturation but agarose films did not. In these studies, the biomaterial adjuvant effect was attenuated by material selection (PLGA or agarose scaffolds) or local delivery of an anti-inflammatory/immunosuppressive glucocorticoid, dexamethasone (DX), from PLGA scaffolds. Porous scaffolds (SCs) of PLGA or agarose were produced to deliver equivalent amounts of model antigen, ovalbumin (OVA). Alternatively, PLGA SCs with incorporated OVA were produced with or without DX. These SCs were implanted individually, subcutaneously, and dorsally in C57BL/6 mice. Blood was collected from mice at specific times over a 12-week duration for measurement of antibody production against OVA. Scaffolds were explanted at 12 weeks for histological examination of foreign body response. Scaffolds of PLGA, but not of agarose, were found to elicit higher antibody production against co-delivered OVA, than negative controls. Short-term delivery of DX from PLGA SCs delivering OVA temporarily delayed onset of anti-OVA antibody production. More sustained release of DX at an effective dose and with an appropriate time course is expected to extend the effect of DX on the biomaterial adjuvant effect. The immunomodulatory ability of biomaterials to affect the immune response to co-delivered antigen is demonstrated wherein this immunomodulatory ability correlates with the observed in vitro differential effects of biomaterials on DC maturation.


Subject(s)
Adjuvants, Immunologic/metabolism , Anti-Inflammatory Agents/administration & dosage , Biocompatible Materials/metabolism , Dexamethasone/administration & dosage , Immunity, Humoral , Lactic Acid/immunology , Adjuvants, Immunologic/chemistry , Animals , Anti-Inflammatory Agents/immunology , Biocompatible Materials/chemistry , Dendritic Cells/immunology , Dexamethasone/immunology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Sepharose/immunology , Tissue Scaffolds/chemistry
16.
J Biomed Mater Res A ; 94(1): 252-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20166218

ABSTRACT

Inflammatory responses to implanted biomedical devices elicit a foreign body fibrotic reaction that limits device integration and performance in various biomedical applications. We examined chronic inflammatory responses to microgel conformal coatings consisting of thin films of poly(N-isopropylacrylamide) hydrogel microparticles cross-linked with poly(ethylene glycol) diacrylate deposited on poly(ethylene terephthalate) (PET). Unmodified and microgel-coated PET disks were implanted subcutaneously in rats for 4 weeks and explants were analyzed by histology and immunohistochemistry. Microgel coatings reduced chronic inflammation and resulted in a more mature/organized fibrous capsule. Microgel-coated samples exhibited 22% thinner fibrous capsules that contained 40% fewer cells compared to unmodified PET disks. Furthermore, microgel-coated samples contained significantly higher levels of macrophages (80%) than unmodified PET controls. These results demonstrate that microgel coatings reduce chronic inflammation to implanted biomaterials. (c) 2010 Wiley Periodicals, Inc. J Biomed Mater Res, 2010.


Subject(s)
Coated Materials, Biocompatible/metabolism , Hydrogels/metabolism , Implants, Experimental , Acrylamides/chemistry , Acrylamides/immunology , Acrylic Resins , Animals , Coated Materials, Biocompatible/chemistry , Foreign-Body Reaction/immunology , Foreign-Body Reaction/pathology , Hydrogels/chemistry , Implants, Experimental/adverse effects , Inflammation , Male , Materials Testing , Polymers/chemistry , Rats , Rats, Wistar
17.
Acta Biomater ; 6(7): 2621-30, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20097314

ABSTRACT

A variety of combination products composed of biomaterials and biologics have been developed for tissue regeneration or vaccine delivery. The host immune response to the immunogenic biological components in such products may be modulated by the biomaterial component. Distinct biomaterials have been shown to differentially affect the maturation of dendritic cells (DCs). DCs are professional antigen-presenting cells (APCs) that bridge innate and adaptive immunity and play a central role in inducing immunity or initiating immune tolerance. However, the biomaterials systems used to study DC response thus far have been insufficient to draw a clear conclusion as to which biomaterial properties are the key to controlling DC phenotype. In this study, we developed a 96-well filter plate-based high-throughput (HTP) methodology to assess DC maturation upon biomaterial treatment. Equivalent biomaterial effects on DC phenotype were measured using the conventional flow cytometric and filter-plate method, which validated the HTP methodology. This methodology will be used to screen a large number of biomaterials simultaneously and to draw correlations between material properties and DC phenotype, thereby providing biomaterial design criteria and immunomodulatory strategies for both tissue engineering and vaccine delivery applications.


Subject(s)
Biocompatible Materials , Dendritic Cells , Humans , Phenotype
18.
Biomaterials ; 31(4): 594-601, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19818491

ABSTRACT

The host response to a biomaterial is characterized by both acute recruitment and attachment of cells as well as chronic encapsulating tissue reaction. The implantation procedure induces production of damage-associated molecular patterns (DAMPs) which may contribute to host recognition of the material. Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) that bind not only pathogen-associated molecular patterns (PAMPs) but also DAMPs. We sought to investigate whether TLR4/DAMP interactions were involved in the acute and chronic inflammatory response to an implanted biomaterial. When PET discs were implanted intraperitoneally for 16h, no differences were found in the number of leukocytes recruited between TLR4(+) (C57BL/10J) and TLR4(-) (C57BL/10ScNJ) mice. However, a significant shift in the leukocyte profile on the biomaterial surface was observed for TLR4(-) mice. While the total number of adherent cells was the same in both strains, TLR4(+) mice had a profile with equivalent neutrophil and monocyte/macrophage presence on the material surface, and TLR4(-) mice had a profile of predominantly neutrophils with fewer monocyte/macrophages. When implants were placed subcutaneously for 2 weeks, the fibrous capsule thicknesses were not different between TLR4(+) and TLR4(-) mouse strains. These findings illustrate that TLR4 may play a role in the initial recognition of a biomaterial by directing the adhesive cellular profile.


Subject(s)
Biocompatible Materials/pharmacology , Leukocytes/cytology , Leukocytes/drug effects , Toll-Like Receptor 4/genetics , Animals , Biocompatible Materials/adverse effects , Enzyme-Linked Immunosorbent Assay , Inflammation/immunology , Inflammation/metabolism , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/cytology , Monocytes/drug effects , Muscles/immunology , Muscles/metabolism , Neutrophils/cytology , Neutrophils/drug effects , Skin/immunology , Skin/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
J Biomed Mater Res A ; 92(2): 791-800, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19274715

ABSTRACT

Macrophages are central mediators of biomaterial-associated wound healing. Dendritic cells (DCs) link innate and adaptive immunity and are important in the context of the host response to combination products. Starting with human peripheral blood mononuclear cells (PBMCs), DCs were derived from monocytes upon culture with granulocyte macrophage colony-stimulating factor and interleukin-4; macrophages were derived from monocytes upon culture without cytokines. Macrophage or DC cultures were characterized at relevant timepoints in both adherent and nonadherent fractions on control Primaria surfaces to characterize and define these inflammatory/immune cells as a prequel to their use in in vitro test biomaterial-host response studies. At day 10 (typical time for harvesting macrophages for subsequent treatment with test biomaterials), macrophages were CD11c+, macrophage mannose receptor (MMR)+, CD14+, and CD64+. At day 6 (typical time for harvesting of DCs after 24-h treatment with test biomaterials), DCs were CD1c+, CD11c+, CD123+, MMR+, CD14+, and CD64-. Furthermore, CD3+ and CD4+ T lymphocytes and CD19+ and CD24+ B lymphocytes were present in both cultures at all timepoints, although to different extents. Immature DCs (approximately 15 microm), were rounded but presented extensive dendritic processes upon maturation with lipopolysaccharide. Alternatively, adherent macrophages (approximately 15-20 microm) displayed internalized lipids and exhibited few membrane processes. The characterization and comparison of existing techniques to establish reliable, reproducible primary cultures of DCs or macrophages provides an important basis for examining and interpreting complex macrophage/DC-lymphocyte-orchestrated host responses in future studies with equivalent cell populations on test biomaterials.


Subject(s)
Biocompatible Materials/pharmacology , Dendritic Cells/physiology , Macrophages/physiology , Materials Testing/methods , Animals , Antigen-Presenting Cells/drug effects , Biomarkers/metabolism , CD4-Positive T-Lymphocytes , Cell Adhesion , Cells, Cultured , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Interleukin-4/pharmacology , Swine
20.
J Biomed Mater Res A ; 92(4): 1487-99, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19425048

ABSTRACT

Biomaterial contact triggers dendritic cell (DC) maturation, to an extent depending on the biomaterial, ultimately enhancing an immune response toward associated antigens, implying a role for biomaterials as adjuvants. Self-assembled monolayers (SAM) of alkanethiols on titanium/gold-coated surfaces presenting different chemistries were used to study effects of biomaterial surface chemistry on DC maturation. Although DCs treated with OH, COOH, or NH(2) SAMs showed modest maturation, those treated with CH(3) SAMs were least mature, all based on cytospins, allostimulatory capacity, or maturation marker expression. Surprisingly, DCs treated with CH(3) SAMs secreted highest levels of proinflammatory tumor necrosis factor-alpha (TNF-alpha) or interleukin-6 (IL-6) but were least mature. Secretion of anti-inflammatory mediators by DCs treated with CH(3) SAMs was not responsible for mitigating DC maturation under these conditions. Interestingly, elevated levels of apoptotic markers were measured associated with DCs and T cells upon CH(3) SAMs contact. Since phagocytosis of apoptotic DCs has strong immunosuppressive effects on DCs, more apoptotic DCs on CH(3) SAMs may account for lower DC maturation. Finally, higher expression of cytotoxic T lymphocyte associated antigen receptor-4 (CTLA-4) on T cells may imply a mechanism of T cell inhibition on CH(3) SAMs.


Subject(s)
Biocompatible Materials , Dendritic Cells/immunology , Adjuvants, Immunologic , Alkanes/chemistry , Antigens, CD/immunology , Apoptosis/physiology , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biomarkers/metabolism , CTLA-4 Antigen , Cell Culture Techniques , Cell Shape , Cells, Cultured , Cytokines/immunology , Dendritic Cells/cytology , Dendritic Cells/physiology , Gold/chemistry , Humans , Interleukin-6/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Materials Testing , Sulfhydryl Compounds/chemistry , Surface Properties , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tissue Engineering/instrumentation , Tissue Engineering/methods , Titanium/chemistry , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...