Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Cell Physiol ; 235(6): 5120-5129, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31674663

RESUMO

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.


Assuntos
Células Dendríticas/metabolismo , Hiperglicemia/metabolismo , Tolerância Imunológica/genética , Linfócitos T Reguladores/imunologia , Animais , Antígeno B7-2/genética , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Diferenciação Celular/imunologia , Células Dendríticas/patologia , Glucose/metabolismo , Humanos , Hiperglicemia/genética , Hiperglicemia/imunologia , Hiperglicemia/patologia , Tolerância Imunológica/imunologia , Interleucina-10/farmacologia , Camundongos , Linfócitos T Reguladores/patologia
2.
Annu Rev Biomed Eng ; 17: 317-49, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26421896

RESUMO

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.


Assuntos
Materiais Biocompatíveis , Imunomodulação , Animais , Células Apresentadoras de Antígenos/imunologia , Células Artificiais/imunologia , Engenharia Biomédica , Células Dendríticas/imunologia , Humanos , Imunização , Fatores Imunológicos/administração & dosagem , Imunoterapia , Tecido Linfoide/imunologia , Nanopartículas , Nanotecnologia , Proteínas/imunologia , Biologia de Sistemas , Engenharia Tecidual
3.
J Biomed Mater Res A ; 112(8): 1317-1336, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38562052

RESUMO

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.


Assuntos
Células Dendríticas , Hidrogéis , Interleucina-10 , Polietilenoglicóis , Polietilenoglicóis/química , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Hidrogéis/química , Interleucina-10/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Humanos
4.
ACS Biomater Sci Eng ; 8(10): 4341-4353, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36134725

RESUMO

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.


Assuntos
Hidrogéis , Interleucina-10 , Materiais Biocompatíveis/farmacologia , Células Dendríticas/metabolismo , Etilenos , Fatores de Transcrição Forkhead/metabolismo , Hidrogéis/química , Hidrogéis/farmacologia , Maleimidas/química , Fenótipo , Polietilenoglicóis/química
5.
J Biomed Mater Res A ; 109(7): 1247-1255, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33040412

RESUMO

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.


Assuntos
Células Dendríticas/transplante , Encefalomielite Autoimune Experimental/terapia , Hidrogéis/química , Esclerose Múltipla/terapia , Alicerces Teciduais/química , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Polietilenoglicóis/química
6.
ACS Biomater Sci Eng ; 6(7): 4062-4076, 2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-33463332

RESUMO

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.


Assuntos
Materiais Biocompatíveis , Células Dendríticas , Teorema de Bayes , Materiais Biocompatíveis/farmacologia , Células Cultivadas , Humanos , Fenótipo
7.
Biomaterials ; 28(25): 3626-31, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17521718

RESUMO

Host responses to biomaterials control the biological performance of implanted medical devices. Upon implantation, synthetic materials adsorb biomolecules, which trigger an inflammatory cascade comprising coagulation, leukocyte recruitment/adhesion, and foreign body reaction. The foreign body reaction and ensuing fibrous encapsulation severely limit the in vivo performance of numerous biomedical devices. While it is well established that plasma fibrinogen and secreted cytokines modulate leukocyte recruitment and maturation into foreign body giant cells, mediators of chronic inflammation and fibrous encapsulation of implanted biomaterials remain poorly understood. Using plasma fibronectin (pFN) conditional knock-out mice, we demonstrate that pFN modulates the foreign body response to polyethylene terephthalate disks implanted subcutaneously. Fibrous collagenous capsules were two-fold thicker in mice depleted of pFN compared to controls. In contrast, deletion of pFN did not alter acute leukocyte recruitment to the biomaterial, indicating that pFN modulates chronic fibrotic responses. The number of foreign body giant cells associated with the implant was three times higher in the absence of pFN while macrophage numbers were not different, suggesting that pFN regulates the formation of biomaterial-associated foreign body giant cells. Interestingly, cellular FN (cFN) was present in the capsules of both normal and pFN-depleted mice, suggesting that cFN could not compensate for the loss of pFN. These results implicate pFN in the host response to implanted materials and identify a potential target for therapeutic intervention to enhance the biological performance of biomedical devices.


Assuntos
Materiais Biocompatíveis/toxicidade , Fibronectinas/fisiologia , Reação a Corpo Estranho/fisiopatologia , Polietilenotereftalatos/toxicidade , Animais , Materiais Biocompatíveis/administração & dosagem , Feminino , Fibronectinas/sangue , Fibronectinas/genética , Reação a Corpo Estranho/induzido quimicamente , Deleção de Genes , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Plasma/química , Polietilenotereftalatos/administração & dosagem , Fatores de Tempo
8.
J Biomed Mater Res A ; 80(1): 7-12, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16958042

RESUMO

Understanding of biomaterial adjuvant effect and its mechanisms is essential for the effective design and selection of appropriate materials for specific applications. We have previously shown that poly(lactic-co-glycolic acid) (PLGA), one of the most commonly studied polymers in tissue engineering, supports an adjuvant effect as measured by enhanced immune response against a co-delivered model antigen, which was dependent on the form of the biomaterial. Furthermore, we have shown that PLGA induces the maturation of human peripheral blood mononuclear cell-derived dendritic cells (DCs) in vitro. In this study, the effect of PLGA contact on the maturation of murine bone marrow-derived DCs was investigated in part to explain the biomaterial adjuvant effect observed. Treatment of bone marrow-derived DCs from C57BL6 mice with PLGA microparticles or films lead to maturation of these cells as exemplified by increased expression of co-stimulatory molecules CD80 and CD86 and production of proinflammatory cytokines TNF-alpha and IL-6. These results suggest that PLGA contact induces maturation of murine DCs, supporting our observations with human DCs. With the techniques developed in this study and given the results, our future goal is to utilize transgenic murine models to delineate the mechanisms of biomaterial-induced DC maturation.


Assuntos
Adjuvantes Imunológicos , Materiais Biocompatíveis , Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Glicolatos/imunologia , Teste de Materiais , Animais , Antígeno B7-2/biossíntese , Antígeno B7-2/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Humanos , Interleucina-6/biossíntese , Interleucina-6/imunologia , Ácido Láctico , Camundongos , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Engenharia Tecidual , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
9.
J Control Release ; 112(2): 271-9, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16549219

RESUMO

Usage of DNA vaccination has been limited by inefficient cellular expression of plasmid constructs used in DNA vaccines. We describe a novel system for enhancing delivery of DNA vaccine plasmids into cells and their nuclei. This delivery system uses recombinant reovirus type 3 sigma1 attachment protein genetically modified with a nuclear localization sequence (sigma1-NLS) as a targeting ligand. Purified sigma1-NLS was covalently conjugated to the polycation polyethyleneimine (PEI) using a carboxyl-reactive cross-linking agent and complexed with plasmid DNA. The benefit of the NLS in enhancement of protein delivery into the nucleus was demonstrated by liposome-mediated loading of cells with sigma1 or sigma1-NLS. In L929 fibroblasts loaded with sigma1-NLS, 69% of the internalized protein was recovered in the nuclear fraction after 6 h compared to just 10% when using unmodified sigma1. Transfection of L929 cells with sigma1-NLS-conjugated PEI complexed with a luciferase expression plasmid resulted in a mean 16-fold increase in luciferase activity over complexes made with unmodified PEI, compared to a mean 3-fold boost obtained using sigma1-conjugated PEI. These results suggest that sigma1-NLS is a useful bifunctional targeting ligand suitable for enhancing DNA delivery and subsequent gene expression for both DNA vaccine applications and nonviral gene therapy.


Assuntos
Proteínas do Capsídeo/genética , Reagentes de Ligações Cruzadas/administração & dosagem , Sinais de Localização Nuclear/genética , Plasmídeos/administração & dosagem , Polietilenoimina/administração & dosagem , Vacinas de DNA , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Luciferases/genética , Orthoreovirus Mamífero 3/genética , Camundongos , Plasmídeos/genética , Proteínas Recombinantes de Fusão/genética , Transfecção
10.
J Biomed Mater Res A ; 79(2): 393-408, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16886225

RESUMO

Application of biomaterials in combination products in which the biomaterial is presented to the host with a biological component prompts the need for understanding the biomaterial-associated adjuvant effect in the immune response against antigens associated with such a product. We have previously demonstrated that a polymer commonly used in tissue engineering and vaccine delivery, poly(lactic-co-glycolic acid) (PLGA), exerts an adjuvant effect in vivo, which was supported by PLGA-induced dendritic cell (DC) maturation in vitro. In this study, the effects of agarose and PLGA on DC maturation were compared in vitro to establish differential biomaterial effects. Human monocyte-derived DCs were treated with agarose or PLGA microparticles or films, and their maturation effect was measured as expression of costimulatory and MHC class II molecules, allostimulatory capacity, and proinflammatory cytokine secretion. Direct comparison of DC maturation phenotype indicated that PLGA was a stronger stimulus of DC maturation than agarose, and this maturation was not affected by microparticle phagocytosis. However, agarose-treated DCs showed higher activation of nuclear factor kappaB (NFkappaB) 24 h after the initial stimulation of DCs. Taken together, these results demonstrate differential biomaterial effects on DC maturation, substantiating the maturation effect of PLGA, and provide screening methods for biomaterial adjuvant effect for applications in combination products.


Assuntos
Materiais Biocompatíveis/química , Técnicas de Cultura de Células/métodos , Células Dendríticas/citologia , Glicolatos/química , Sefarose/química , Cromatografia Gasosa , Células Dendríticas/metabolismo , Humanos , Ácido Láctico , Espectrometria de Massas , Microscopia Eletrônica de Varredura , Microesferas , Fagocitose , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/química , Temperatura , Engenharia Tecidual , Fator de Necrose Tumoral alfa/metabolismo
11.
Biomaterials ; 26(16): 2991-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15603794

RESUMO

A model shed antigen, ovalbumin (OVA), was incorporated into polymeric biomaterial carriers made of poly(lactic-co-glycolic acid) (PLGA) in the form of microparticles (MP) or scaffolds (SC). These polymeric biomaterial carrier vehicles with incorporated antigen were then injected or implanted into mice and the resulting time-dependent systemic humoral immune response towards the controlled released OVA was assessed by following the OVA-specific IgG concentration and isotypes using ELISA. To assess the differential level of enhancement of the immune response depending on the form of carrier vehicle (MP vs. SC), the total amount of polymer and OVA delivered was kept constant as well as the release rate of OVA for both carrier vehicles. The level of the humoral immune response was higher and sustained for OVA released from PLGA SC which were implanted with associated tissue damage, and lower and transient when the same amount of polymer and OVA were delivered from PLGA MP, which were minimally invasively delivered by injection. This immune response was primarily Th2 helper T cell-dependent, although for the strong adjuvant, CFA, and PLGA SC carriers there was both a Th2 and Th1 response contribution. These results implicate 'danger signals' associated with the implantation of the scaffolds due to tissue injury which primed the system for an enhanced immune response.


Assuntos
Materiais Biocompatíveis/química , Glicolatos/química , Ovalbumina/química , Engenharia Tecidual/métodos , Animais , Antígenos/química , Portadores de Fármacos/química , Ensaio de Imunoadsorção Enzimática , Sistema Imunitário/imunologia , Imunoglobulina G/química , Cinética , Ácido Láctico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/química , Linfócitos T/imunologia , Células Th1/imunologia , Células Th2/imunologia , Fatores de Tempo
12.
J Biomed Mater Res A ; 72(3): 269-78, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15657946

RESUMO

Understanding endothelial cell (EC)/blood/biomaterial interactions is crucial for the advancement of cardiovascular devices that often fail because of the lack of nonthrombogenic biomaterials. To begin to assess these interactions, a static EC/blood cell/biomaterial model was used. Isolated blood cells were pretreated with model biomaterial beads with different surface chemistries: polystyrene (PS), and PS beads grafted with 3-kDa polyethylene glycol (PEG) with either a hydroxyl (PS-PEG-OH) or amine (PS-PEG-NH(2)) terminal group at 5.4 or 54 x 10(4) beads/mL. Biomaterial-treated monocytes, neutrophils, or platelets were applied to human umbilical vein ECs (HUVECs) for 5 or 24 h of static coculture, and the resultant procoagulant HUVEC phenotype was characterized using several methods. Flow cytometry was used to assess surface expression of tissue factor (TF), adenosine triphosphate diphosphohydrolase, phosphatidylserine, and thrombomodulin, a functional TF assay was used to assess TF activity, and a plasma recalcification assay examined clotting times on HUVECs. Static coculture of HUVEC with biomaterial-treated neutrophils induced a procoagulant phenotype as exemplified by upregulation of TF expression and total functional activity, and downregulation of adenosine triphosphate diphosphohydrolase and thrombomodulin expression. The plasma recalcification assay demonstrated that HUVECs cocultured with biomaterial-treated monocytes significantly shortened clotting times, with some effect of similarly treated neutrophils.


Assuntos
Materiais Biocompatíveis , Coagulação Sanguínea/fisiologia , Células Endoteliais/fisiologia , Monócitos/fisiologia , Biomarcadores , Técnicas de Cocultura , Humanos , Cinética , Plasma/metabolismo , Fatores de Tempo , Veias Umbilicais/fisiologia
13.
J Biomed Mater Res A ; 74(4): 503-10, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16158496

RESUMO

Immature dendritic cells (iDCs) were derived from human peripheral blood monocytes, and treated with films of biomaterials commonly used in combination products (e.g., tissue engineered constructs or vaccines) to assess the resultant dendritic cell (DC) maturation compared to positive control of lipopolysaccharide (LPS) treatment for DC maturation or negative control of untreated iDCs. The following biomaterials were tested: alginate, agarose, chitosan, hyaluronic acid, 75:25 poly(lactic-co-glycolic acid) (PLGA). The effect of DC culture on these films was undertaken to identify biomaterials which support DC maturation and those biomaterials that did not. Dendritic cells treated with chitosan or PLGA (agarose to a lesser extent) films increased expression levels of CD86, CD40, and HLA-DQ, compared to control iDCs, similar to LPS-matured DCs, whereas DCs treated with alginate or hyaluronic acid films decreased their expression levels of these same molecules. In summary, a differential effect of the biomaterial on which iDCs were cultured was observed as far as the extent of induced DC maturation. The effect of biomaterials on DC maturation, and the associated adjuvant effect, is a novel biocompatibility selection and design criteria for biomaterials to be used in combination products in which immune consequences are potential complications or outcomes.


Assuntos
Materiais Biocompatíveis , Diferenciação Celular , Células Dendríticas/fisiologia , Teste de Materiais , Monócitos/fisiologia , Antígenos CD/biossíntese , Antígeno B7-2 , Antígenos CD40/biossíntese , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/citologia , Humanos , Lipopolissacarídeos/farmacologia , Teste de Materiais/métodos , Glicoproteínas de Membrana/biossíntese , Monócitos/citologia
14.
J Biomed Mater Res A ; 103(1): 170-84, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24616366

RESUMO

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.


Assuntos
Materiais Biocompatíveis , Células Dendríticas/citologia , Imunofenotipagem , Linfócitos T/citologia , Biomarcadores/metabolismo , Polaridade Celular , Técnicas de Cocultura , Células Dendríticas/imunologia , Humanos
15.
Biomaterials ; 25(2): 295-304, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14585717

RESUMO

A model shed antigen, ovalbumin (OVA), was co-delivered with polymeric biomaterial carrier vehicles in C57BL6 mice to test whether the presence of the biomaterial acted as an adjuvant in the immune response towards the associated antigen. The biomaterials tested were non-biodegradable polystyrene microparticles and biodegradable 50:50 or 75:25 poly(lactic-co-glycolic acid) (PLGA) microparticles or scaffolds. For each biomaterial carrier vehicle, to assess the resulting time-dependent systemic humoral immune response towards the co-delivered OVA, the OVA-specific IgG concentration and isotypes (IgG2a or IgG1, indicating a predominant Th1 or Th2 response, respectively) were determined using ELISA. OVA co-delivered with biomaterial carrier vehicles supported a moderate humoral immune response that was maintained for the 18-week duration of the experiment. This humoral immune response was primarily Th2 helper T cell-dependent as indicated by the predominant IgG1 isotype. Furthermore, this humoral immune response was not material chemistry-dependent within the material set tested here. With the presence of the biomaterial resulting in an enhancement of the humoral immune response to co-delivered antigen, it appears that the biomaterial acts as an adjuvant in the development of an adaptive immune response to co-delivered antigen.


Assuntos
Formação de Anticorpos/imunologia , Materiais Biocompatíveis/metabolismo , Ovalbumina/imunologia , Engenharia Tecidual , Animais , Formação de Anticorpos/fisiologia , Masculino , Camundongos , Ovalbumina/metabolismo
16.
J Biomed Mater Res A ; 64(3): 397-410, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12870471

RESUMO

An understanding of the endothelial cell/blood/biomaterial interactions is central to advancing the success of cardiovascular devices that continue to fail because of the lack of nonthrombogenic biomaterials. A simplified endothelial cell/blood cell/biomaterial static model was used to assess these interactions. Human whole blood or isolated blood cells (mononuclear cells, neutrophils, platelets) were pretreated with biomaterial beads with different surface chemistries: polystyrene (PS), PS beads grafted with 3-kDa polyethylene glycol (PEG) with either hydroxyl (PS-PEG-OH) or amine (PS-PEG-NH2) terminal groups at bead concentrations of 5.4 or 54 x 10(4) beads/mL. Leukocyte and platelet activation and microparticle formation was assessed using flow cytometry. Biomaterial-activated whole blood or isolated cells or mononuclear cell fractions were applied to human umbilical cord endothelial cells (HUVEC) for static coculture, and the resultant proinflammatory HUVEC phenotype was characterized. ICAM-1 and E-selectin expression on HUVEC was increased after 4-h static coculture with biomaterial-treated human whole blood or mononuclear cells but not neutrophils or platelets. VCAM-1 expression on HUVEC was similarly increased after 24-h static coculture but not after 4 h of coculture. Increased concentrations of cytokines, IL-6, IL-8, and MCP-1, were detected in the supernatant of cocultures of HUVEC with biomaterial-treated whole blood or mononuclear cells but not neutrophils or platelets, compared with the media control. After 24 h, cytokine release was significantly increased for both IL-8 and MCP-1 but not IL-6 above concentrations after 4 h of coculture. Neither the cell adhesion molecule (CAM) expression nor cytokine release induced by coculture with biomaterial-treated whole blood or isolated cells was dependent on either material surface chemistry or material surface area. The changes in HUVEC CAM expression and cytokine release induced by biomaterial-treated mononuclear cells can be attributed predominantly to adherent cells on beads and nonadherent bulk cells with moderate regulation by the soluble supernatant; however, mononuclear cell-derived microparticles induced no significant changes in CAM expression or cytokine release after static coculture with HUVEC.


Assuntos
Materiais Biocompatíveis/química , Células Sanguíneas/metabolismo , Endotélio Vascular/citologia , Inflamação/metabolismo , Células Sanguíneas/citologia , Plaquetas/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Selectina E/metabolismo , Endotélio Vascular/metabolismo , Humanos , Ativação Linfocitária , Fenótipo , Ativação Plaquetária , Polietilenoglicóis/química , Poliestirenos/química , Propriedades de Superfície
17.
J Biomed Mater Res A ; 71(1): 45-54, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15368253

RESUMO

Immature dendritic cells (iDCs) were derived from human peripheral blood monocytes, and treated with 75:25 poly(lactic-co-glycolic acid) (PLGA) microparticles (MPs) or film to assess the resultant dendritic cell (DC) maturation as compared to positive control of lipopolysaccharide (LPS) treatment for DC maturation or negative control of untreated iDCs. The effect of PLGA contact on DC maturation was examined as one possible explanation for the PLGA adjuvant effect we have observed in the enhancement of an immune response to codelivered model antigen, as adjuvants act through the maturation of DCs. Culturing iDCs with PLGA MPs or PLGA film resulted in morphology similar to that of LPS-matured DCs and the association, or possible internalization, of PLGA MPs. Furthermore, biomaterial-treated iDCs demonstrated an increase in MHC class II and costimulatory molecule expression compared to iDCs but to a lower level than that of LPS-matured DCs. Direct iDC contact with PLGA MPs was necessary for maturation. Immature DCs exposed to PLGA MPs were stimulatory of allogeneic T-cell proliferation, whereas cells exposed to PLGA film were not. Further, PLGA MPs supported a moderate delayed type hypersensitivity reaction in mice indicative of in vivo DC maturation. Taken together, these results suggest that PLGA is a DC maturation stimulus and that the form of the biomaterial may influence the extent of DC maturation.


Assuntos
Materiais Biocompatíveis/farmacologia , Células Dendríticas/efeitos dos fármacos , Glicolatos/farmacologia , Monócitos/efeitos dos fármacos , Animais , Antígenos CD/imunologia , Biomarcadores , Técnicas de Cultura de Células/métodos , Forma Celular , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/fisiologia , Glicolatos/imunologia , Humanos , Hipersensibilidade Tardia , Ácido Láctico , Teste de Cultura Mista de Linfócitos , Teste de Materiais , Camundongos , Monócitos/citologia , Monócitos/imunologia , Monócitos/fisiologia , Fenótipo , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
18.
Biomaterials ; 35(22): 5862-74, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24746228

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Glicoconjugados/química , Glicoconjugados/imunologia , Adsorção , Sobrevivência Celular , Células Cultivadas , Células Dendríticas/citologia , Humanos , Imunidade Inata
19.
Acta Biomater ; 8(10): 3606-17, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22705044

RESUMO

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.


Assuntos
Materiais Biocompatíveis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Quitosana/farmacologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Fluorescência , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Ácido Hialurônico/farmacologia , Mediadores da Inflamação/metabolismo , Ácido Láctico/farmacologia , Lipopolissacarídeos/farmacologia , Espectroscopia Fotoeletrônica , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptores de Superfície Celular/metabolismo , Sefarose/farmacologia , Propriedades de Superfície/efeitos dos fármacos , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo
20.
Biomaterials ; 33(6): 1699-713, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22136715

RESUMO

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.


Assuntos
Materiais Biocompatíveis/química , Materiais Revestidos Biocompatíveis/química , Células Dendríticas/citologia , Ácidos Polimetacrílicos/química , Técnicas de Química Combinatória , Humanos , Cinética , Teste de Materiais , Modelos Químicos , Análise Multivariada , Fenótipo , Polímeros/química , Análise de Componente Principal , Análise de Regressão , Propriedades de Superfície , Temperatura , Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA