Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(43): e2205417119, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36256820

RESUMO

Antigen-specific therapies hold promise for treating autoimmune diseases such as multiple sclerosis while avoiding the deleterious side effects of systemic immune suppression due to delivering the disease-specific antigen as part of the treatment. In this study, an antigen-specific dual-sized microparticle (dMP) treatment reversed hind limb paralysis when administered in mice with advanced experimental autoimmune encephalomyelitis (EAE). Treatment reduced central nervous system (CNS) immune cell infiltration, demyelination, and inflammatory cytokine levels. Mechanistic insights using single-cell RNA sequencing showed that treatment impacted the MHC II antigen presentation pathway in dendritic cells, macrophages, B cells, and microglia, not only in the draining lymph nodes but also strikingly in the spinal cord. CD74 and cathepsin S were among the common genes down-regulated in most antigen presenting cell (APC) clusters, with B cells also having numerous MHC II genes reduced. Efficacy of the treatment diminished when B cells were absent, suggesting their impact in this therapy, in concert with other immune populations. Activation and inflammation were reduced in both APCs and T cells. This promising antigen-specific therapeutic approach advantageously engaged essential components of both innate and adaptive autoimmune responses and capably reversed paralysis in advanced EAE without the use of a broad immunosuppressant.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Camundongos , Antígenos , Citocinas , Camundongos Endogâmicos C57BL , Paralisia , Catepsinas , Imunossupressores/uso terapêutico
2.
Semin Immunol ; 29: 33-40, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28487131

RESUMO

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.


Assuntos
Materiais Biocompatíveis/metabolismo , Células Dendríticas/imunologia , Próteses e Implantes , Imunidade Adaptativa , Animais , Reação a Corpo Estranho , Humanos , Imunidade Inata , Medicina Regenerativa , Engenharia Tecidual
3.
Bioconjug Chem ; 29(3): 649-656, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29285931

RESUMO

Enzymes are attractive as immunotherapeutics because they can catalyze shifts in the local availability of immunostimulatory and immunosuppressive signals. Clinical success of enzyme immunotherapeutics frequently hinges upon achieving sustained biocatalysis over relevant time scales. The time scale and location of biocatalysis are often dictated by the location of the substrate. For example, therapeutic enzymes that convert substrates distributed systemically are typically designed to have a long half-life in circulation, whereas enzymes that convert substrates localized to a specific tissue or cell population can be more effective when designed to accumulate at the target site. This Topical Review surveys approaches to improve enzyme immunotherapeutic efficacy via chemical modification, encapsulation, and immobilization that increases enzyme accumulation at target sites or extends enzyme half-life in circulation. Examples provided illustrate "replacement therapies" to restore deficient enzyme function, as well as "enhancement therapies" that augment native enzyme function via supraphysiologic doses. Existing FDA-approved enzyme immunotherapies are highlighted, followed by discussion of emerging experimental strategies such as those designed to enhance antitumor immunity or resolve inflammation.


Assuntos
Terapia Enzimática/métodos , Imunoterapia/métodos , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/uso terapêutico , Antineoplásicos/química , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Asparaginase/química , Asparaginase/imunologia , Asparaginase/uso terapêutico , Biocatálise , Enzimas Imobilizadas/química , Enzimas Imobilizadas/imunologia , Enzimas Imobilizadas/uso terapêutico , Doença de Fabry/imunologia , Doença de Fabry/terapia , Doença de Gaucher/imunologia , Doença de Gaucher/terapia , Glucosilceramidase/química , Glucosilceramidase/imunologia , Glucosilceramidase/uso terapêutico , Glicosilação , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/uso terapêutico , Inflamação/imunologia , Inflamação/terapia , Doenças por Armazenamento dos Lisossomos/imunologia , Doenças por Armazenamento dos Lisossomos/terapia , Neoplasias/imunologia , Neoplasias/terapia , alfa-Galactosidase/química , alfa-Galactosidase/imunologia , alfa-Galactosidase/uso terapêutico
4.
Proc Natl Acad Sci U S A ; 112(28): 8732-7, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26124098

RESUMO

A new paradigm in oncology establishes a spectrum of tumorigenic potential across the heterogeneous phenotypes within a tumor. The cancer stem cell hypothesis postulates that a minute fraction of cells within a tumor, termed cancer stem cells (CSCs), have a tumor-initiating capacity that propels tumor growth. An application of this discovery is to target this critical cell population using chemotherapy; however, the process of isolating these cells is arduous, and the rarity of CSCs makes it difficult to test potential drug candidates in a robust fashion, particularly for individual patients. To address the challenge of screening drug libraries on patient-derived populations of rare cells, such as CSCs, we have developed a drug-eluting microarray, a miniaturized platform onto which a minimal quantity of cells can adhere and be exposed to unique treatment conditions. Hundreds of drug-loaded polymer islands acting as drug depots colocalized with adherent cells are surrounded by a nonfouling background, creating isolated culture environments on a solid substrate. Significant results can be obtained by testing <6% of the cells required for a typical 96-well plate. Reliability was demonstrated by an average coefficient of variation of 14% between all of the microarrays and 13% between identical conditions within a single microarray. Using the drug-eluting array, colorectal CSCs isolated from two patients exhibited unique responses to drug combinations when cultured on the drug-eluting microarray, highlighting the potential as a prognostic tool to identify personalized chemotherapeutic regimens targeting CSCs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Células-Tronco Neoplásicas/efeitos dos fármacos , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/patologia , Humanos , Pessoa de Meia-Idade , Células Tumorais Cultivadas
5.
Clin Immunol ; 160(1): 90-102, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25842187

RESUMO

We developed a novel poly(lactic-co-glycolic acid)-based, microparticle (MP) system providing concurrent delivery of multiple encapsulated immuno-suppressive factors and antigen, for in vivo conditioning of dendritic cells (DCs) toward a tolerance promoting pathway. Subcutaneous administration prevents onset of type 1 diabetes (T1D) in NOD mice. Two MP sizes were made: phagocytosable MPs were fabricated encapsulating vitamin D3 or insulin B(9-23) peptide, while unphagocytosable MPs were fabricated encapsulating TGF-ß1 or GM-CSF. The combination of Vit D3/TGF-ß1 MPs confers an immature and LPS activation-resistant phenotype to DCs, and MP-delivered antigen is efficiently and functionally presented. Notably, two subcutaneous injections into 4week old NOD mice using the combination of MPs encapsulating Vit D3, Ins B, TGF-ß1 and GM-CSF protected 40% of mice from T1D development, significant in comparison to the control. This work represents one of the first applications of a biomaterial-based, MP vaccine system to successfully prevent autoimmune diabetes.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Portadores de Fármacos , Ácido Láctico , Ácido Poliglicólico , Vacinas/administração & dosagem , Animais , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Colecalciferol/farmacologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Tolerância Imunológica/imunologia , Insulina/farmacologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Tamanho da Partícula , Fragmentos de Peptídeos/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Fator de Crescimento Transformador beta1/farmacologia , Vacinas/imunologia
6.
Angiogenesis ; 17(4): 823-830, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24957885

RESUMO

Arteriovenous malformation (AVM) refers to a vascular anomaly where arteries and veins are directly connected through a complex, tangled web of abnormal AV fistulae without a normal capillary network. Hereditary hemorrhagic telangiectasia (HHT) types 1 and 2 arise from heterozygous mutations in endoglin (ENG) and activin receptor-like kinase 1 (ALK1), respectively. HHT patients possess AVMs in various organs, and telangiectases (small AVMs) along the mucocutaneous surface. Understanding why and how AVMs develop is crucial for developing therapies to inhibit the formation, growth, or maintenance of AVMs in HHT patients. Previously, we have shown that secondary factors such as wounding are required for Alk1-deficient vessels to develop skin AVMs. Here, we present evidences that AVMs establish from nascent arteries and veins rather than from remodeling of a preexistent capillary network in the wound-induced skin AVM model. We also show that VEGF can mimic the wound effect on skin AVM formation, and VEGF-neutralizing antibody can prevent skin AVM formation and ameliorate internal bleeding in Alk1-deficient adult mice. With topical applications at different stages of AVM development, we demonstrate that the VEGF blockade can prevent the formation of AVM and cease the progression of AVM development. Taken together, the presented experimental model is an invaluable system for precise molecular mechanism of action of VEGF blockades as well as for preclinical screening of drug candidates for epistaxis and gastrointestinal bleedings.


Assuntos
Malformações Arteriovenosas/metabolismo , Telangiectasia Hemorrágica Hereditária/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/farmacologia , Encéfalo/patologia , Modelos Animais de Doenças , Progressão da Doença , Heterozigoto , Camundongos , Camundongos Knockout , Mutação , Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
7.
MRS Bull ; 39(1): 25-34, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26997752

RESUMO

Recently, biomaterial scientists have married materials engineering and immunobiology to conceptualize new immunomodulatory materials. This special class of biomaterials can modulate and harness the innate properties of immune functionality for enhanced therapeutic efficacy. Generally, two fundamental strategies are followed in the design of immunomodulatory biomaterials: (1) immuno-evasive (immuno-mimetic, immuno-suppressing, or immuno-inert) biomaterials and (2) immuno-activating or immuno-enhancing biomaterials. This article highlights the development and application of a number of immunomodulatory materials, categorized by these two general approaches.

8.
Biomaterials ; 294: 122001, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36716589

RESUMO

Antigen-specific therapies allow for modulation of the immune system in a disease relevant context without systemic immune suppression. These therapies are especially valuable in autoimmune diseases such as multiple sclerosis (MS), where autoreactive T cells destroy myelin sheath. This work shows that an antigen-specific dual-sized microparticle (dMP) system can effectively halt and reverse disease progression in a mouse model of MS. Current MS treatments leave patients immunocompromised, but the dMP formulation spares the immune system as mice can successfully clear a Listeria Monocytogenes infection. Furthermore, we highlight design principles for particle based immunotherapies including the importance of delivering factors specific for immune cell recruitment (GM-CSF or SDF-1), differentiation (GM-CSF or FLT3L) and suppression (TGF-ß or VD3) in conjunction with disease relevant antigen, as the entire formulation is required for maximum efficacy. Lastly, the dMP scheme relies on formulating phagocytosable and non-phagocytosable MP sizes to direct payload to target either cell surface receptors or intracellular targets, as the reverse sized dMP formulation failed to reverse paralysis. We also challenge the design principles of the dMP system showing that the size of the MPs impact efficacy and that GM-CSF plays two distinct roles and that both of these must be replaced to match the primary effect of the dMP system. Overall, this work shows the versatile nature of the dMP system and expands the knowledge in particle science by emphasizing design tenets to guide the next generation of particle based immunotherapies.


Assuntos
Doenças Autoimunes , Encefalomielite Autoimune Experimental , Esclerose Múltipla , Camundongos , Animais , Esclerose Múltipla/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Antígenos , Linfócitos T
9.
J Biomed Mater Res A ; 111(2): 185-197, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36082558

RESUMO

Type 1 diabetes (T1D) presents with two therapeutic challenges: the need to correct underlying autoimmunity and restore ß-cell mass. We harnessed the unique capacity of regulatory T cells (Tregs) and the T cell receptor (TCR) to direct tolerance induction along with tissue-localized delivery of therapeutic agents to restore endogenous ß-cell function. Specifically, we designed a combinatorial therapy involving biomaterials-based poly(lactic-co-glycolic acid) nanoparticles co-loaded with the Treg growth factor, IL-2, and the ß-cell regenerative agent, harmine (a tyrosine-regulated kinase 1A [DYRK1A] inhibitor), conjugated to the surface of Tregs. We observed continuous elution of IL-2 and harmine from nanoparticles for at least 7 days in vitro. When conjugated to primary human Tregs, IL-2 nanoparticles provided sufficient IL-2 receptor signaling to support STAT5 phosphorylation for sustained phenotypic stability and viability in culture. Inclusion of poly-L-lysine (PLL) during nanoparticle-cell coupling dramatically increased conjugation efficiency, providing sufficient IL-2 to support in vitro proliferation of IL-2-dependent CTLL-2 cells and primary murine Tregs. In 12-week-old female non-obese diabetic mice, adoptive transfer of IL-2/harmine nanoparticle-conjugated NOD.BDC2.5 Tregs, which express an islet antigen-specific TCR, significantly prevented diabetes demonstrating preserved in vivo viability. These data provide the preclinical basis to develop a biomaterials-optimized cellular therapy to restore immune tolerance and promote ß-cell proliferation in T1D through receptor-targeted drug delivery within pancreatic islets.


Assuntos
Materiais Biocompatíveis , Diabetes Mellitus Experimental , Humanos , Feminino , Animais , Camundongos , Camundongos Endogâmicos NOD , Materiais Biocompatíveis/farmacologia , Linfócitos T Reguladores , Diabetes Mellitus Experimental/tratamento farmacológico , Interleucina-2/farmacologia
10.
Arthritis Res Ther ; 25(1): 173, 2023 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-37723593

RESUMO

OBJECTIVE: Osteoarthritis (OA) is driven by low-grade inflammation, and controlling local inflammation may offer symptomatic relief. Here, we developed an indoleamine 2,3-dioxygenase and galectin-3 fusion protein (IDO-Gal3), where IDO increases the production of local anti-inflammatory metabolites and Gal3 binds carbohydrates to extend IDO's joint residence time. In this study, we evaluated IDO-Gal3's ability to alter OA-associated inflammation and pain-related behaviors in a rat model of established knee OA. METHODS: Joint residence was first evaluated with an analog Gal3 fusion protein (NanoLuc™ and Gal3, NL-Gal3) that produces luminescence from furimazine. OA was induced in male Lewis rats via a medial collateral ligament and medial meniscus transection (MCLT + MMT). At 8 weeks, NL or NL-Gal3 were injected intra-articularly (n = 8 per group), and bioluminescence was tracked for 4 weeks. Next, IDO-Gal3s's ability to modulate OA pain and inflammation was assessed. Again, OA was induced via MCLT + MMT in male Lewis rats, with IDO-Gal3 or saline injected into OA-affected knees at 8 weeks post-surgery (n = 7 per group). Gait and tactile sensitivity were then assessed weekly. At 12 weeks, intra-articular levels of IL6, CCL2, and CTXII were assessed. RESULTS: The Gal3 fusion increased joint residence in OA and contralateral knees (p < 0.0001). In OA-affected animals, both saline and IDO-Gal3 improved tactile sensitivity (p = 0.008), but IDO-Gal3 also increased walking velocities (p ≤ 0.033) and improved vertical ground reaction forces (p ≤ 0.04). Finally, IDO-Gal3 decreased intra-articular IL6 levels within the OA-affected joint (p = 0.0025). CONCLUSION: Intra-articular IDO-Gal3 delivery provided long-term modulation of joint inflammation and pain-related behaviors in rats with established OA.


Assuntos
Galectina 3 , Osteoartrite do Joelho , Masculino , Animais , Ratos , Ratos Endogâmicos Lew , Indolamina-Pirrol 2,3,-Dioxigenase , Interleucina-6 , Inflamação
11.
Res Sq ; 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37131836

RESUMO

Objective : Controlling joint inflammation can improve osteoarthritis (OA) symptoms; however, current treatments often fail to provide long-term effects. We have developed an indoleamine 2,3-dioxygenase and galectin-3 fusion protein (IDO-Gal3). IDO converts tryptophan to kynurenines, directing the local environment toward an anti-inflammatory state; Gal3 binds carbohydrates and extends IDO's joint residence time. In this study, we evaluated IDO-Gal3's ability to alter OA-associated inflammation and pain-related behaviors in a rat model of established knee OA. Methods : Joint residence was first evaluated with an analog Gal3 fusion protein (NanoLuc™ and Gal3, NL-Gal3) that produces luminescence from furimazine. OA was induced in male Lewis rats via a medial collateral ligament and medial meniscus transection (MCLT+MMT). At 8 weeks, NL or NL-Gal3 were injected intra-articularly (n=8 per group), and bioluminescence was tracked for 4 weeks. Next, IDO-Gal3's ability to modulate OA pain and inflammation was assessed. Again, OA was induced via MCLT+MMT in male Lewis rats, with IDO-Gal3 or saline injected into OA-affected knees at 8 weeks post-surgery (n=7 per group). Gait and tactile sensitivity were then assessed weekly. At 12 weeks, intra-articular levels of IL6, CCL2, and CTXII were assessed. Results : The Gal3 fusion increased joint residence in OA and contralateral knees (p<0.0001). In OA-affected animals, IDO-Gal3 improved tactile sensitivity (p=0.002), increased walking velocities (p≤0.033), and improved vertical ground reaction forces (p≤0.04). Finally, IDO-Gal3 decreased intra-articular IL6 levels within the OA-affected joint (p=0.0025). Conclusion : Intra-articular IDO-Gal3 delivery provided long-term modulation of joint inflammation and pain-related behaviors in rats with established OA.

12.
Nat Biomed Eng ; 7(9): 1156-1169, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37127708

RESUMO

The treatment of chronic inflammation with systemically administered anti-inflammatory treatments is associated with moderate-to-severe side effects, and the efficacy of locally administered drugs is short-lived. Here we show that inflammation can be locally suppressed by a fusion protein of the immunosuppressive enzyme indoleamine 2,3-dioxygenase 1 (IDO) and galectin-3 (Gal3). Gal3 anchors IDO to tissue, limiting the diffusion of IDO-Gal3 away from the injection site. In rodent models of endotoxin-induced inflammation, psoriasis, periodontal disease and osteoarthritis, the fusion protein remained in the inflamed tissues and joints for about 1 week after injection, and the amelioration of local inflammation, disease progression and inflammatory pain in the animals were concomitant with homoeostatic preservation of the tissues and with the absence of global immune suppression. IDO-Gal3 may serve as an immunomodulatory enzyme for the control of focal inflammation in other inflammatory conditions.


Assuntos
Galectina 2 , Indolamina-Pirrol 2,3,-Dioxigenase , Animais , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Progressão da Doença
13.
Sci Immunol ; 8(82): eabn0484, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37115913

RESUMO

The networks of transcription factors (TFs) that control intestinal-resident memory CD8+ T (TRM) cells, including multipotency and effector programs, are poorly understood. In this work, we investigated the role of the TF Bcl11b in TRM cells during infection with Listeria monocytogenes using mice with post-activation, conditional deletion of Bcl11b in CD8+ T cells. Conditional deletion of Bcl11b resulted in increased numbers of intestinal TRM cells and their precursors as well as decreased splenic effector and circulating memory cells and precursors. Loss of circulating memory cells was in part due to increased intestinal homing of Bcl11b-/- circulating precursors, with no major alterations in their programs. Bcl11b-/- TRM cells had altered transcriptional programs, with diminished expression of multipotent/multifunctional (MP/MF) program genes, including Tcf7, and up-regulation of the effector program genes, including Prdm1. Bcl11b also limits the expression of Ahr, another TF with a role in intestinal CD8+ TRM cell differentiation. Deregulation of TRM programs translated into a poor recall response despite TRM cell accumulation in the intestine. Reduced expression of MP/MF program genes in Bcl11b-/- TRM cells was linked to decreased chromatin accessibility and a reduction in activating histone marks at these loci. In contrast, the effector program genes displayed increased activating epigenetic status. These findings demonstrate that Bcl11b is a frontrunner in the tissue residency program of intestinal memory cells upstream of Tcf1 and Blimp1, promoting multipotency and restricting the effector program.


Assuntos
Linfócitos T CD8-Positivos , Fatores de Transcrição , Camundongos , Animais , Linfócitos T CD8-Positivos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Diferenciação Celular , Intestinos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
14.
J Nanosci Nanotechnol ; 12(9): 7132-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23035444

RESUMO

Antibacterial coating approaches are being investigated to modify implants to reduce bacterial adhesion and viability in order to reduce implant-associated infection. Nanostructured materials possess unique surface properties, and nanotopographic surfaces have been reported to modulate bacterial adhesion. Zinc oxide (ZnO) films presenting well-controlled nanorod surface structures have recently been developed. To assess the efficacy of ZnO nanorod surfaces as an anti-bacterial coating, we evaluated bacterial adhesion and viability, compared to sputtered ZnO substrates (a relatively flat control) and glass substrates (as a reference). Common implant-associated pathogens, Pseudomonas aeruginosa and Staphylococcus epidermidis were investigated. The number of adherent P. aeruginosa on ZnO nanorod surfaces was found to be reduced compared to glass and sputtered ZnO, while the adherent number of S. epidermidis on the ZnO nanorods was equivalent to glass. Regarding bacteria viability, the ZnO nanorod and sputtered ZnO surfaces demonstrated a modest, but significant bactericidal effect on adherent P. aeruginosa, killing 2.5-fold and 1.7-fold more over the number of dead P. aeruginosa on glass, respectively. A greater bactericidal effect of ZnO substrates on S. epidermidis was found, with sputtered ZnO and ZnO nanorod substrates killing -20-fold and 30-fold more over the number of dead S. epidermidis on glass, respectively. These data support the further investigation and optimization of ZnO nanorod coatings with potential for bacterial adhesion resistance and bactericidal properties.


Assuntos
Antibacterianos/farmacologia , Nanotubos , Óxido de Zinco/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Fluorescência , Microscopia Eletrônica de Varredura
15.
Mol Aspects Med ; 83: 100992, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34332772

RESUMO

Productive engagement of the immune system is a persistent challenge for biomaterials scientists. Immune engineering offers a new perspective on biomaterial design, with immune cell interaction to modulate effector functions at the center. The effector functions of these cells are intimately linked to their metabolic needs and programming. Immune cell metabolism has received renewed attention in recent years, and with each new discovery there is opportunity for biomaterials scientists. This prospectus aims to provide an overview of the most recent advances in biomaterial engagement of immune cells alongside interrogation of immunometabolism, while looking to future avenues of coalescence. Four cell types are highlighted here: neutrophils, macrophages, dendritic cells, and T cells. Consideration of these two fields, and the tools within each, with a forward-looking mindset is the key to a new era of biomaterials.


Assuntos
Materiais Biocompatíveis , Macrófagos , Materiais Biocompatíveis/metabolismo , Comunicação Celular , Humanos , Macrófagos/metabolismo , Neutrófilos
16.
Lab Chip ; 21(18): 3598-3613, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34346460

RESUMO

Microarrays, miniaturized platforms used for high-content studies, provide potential advantages over traditional in vitro investigation in terms of time, cost, and parallel analyses. Recently, microarrays have been leveraged to investigate immune cell biology by providing a platform with which to systematically investigate the effects of various agents on a wide variety of cellular processes, including those giving rise to immune regulation for application toward curtailing autoimmunity. A specific embodiment incorporates dendritic cells cultured on microarrays containing biodegradable microparticles. Such an approach allows immune cell and microparticle co-localization and release of compounds on small, isolated populations of cells, enabling a quick, convenient method to quantify a variety of cellular responses in parallel. In this study, the microparticle microarray platform was utilized to investigate a small library of sixteen generally regarded as safe (GRAS) compounds (ascorbic acid, aspirin, capsaicin, celastrol, curcumin, epigallocatechin-3-gallate, ergosterol, hemin, hydrocortisone, indomethacin, menadione, naproxen, resveratrol, retinoic acid, α-tocopherol, vitamin D3) for their ability to induce suppressive phenotypes in murine dendritic cells. Two complementary tolerogenic index ranking systems were proposed to summarize dendritic cell responses and suggested several lead compounds (celastrol, ergosterol, vitamin D3) and two secondary compounds (hemin, capsaicin), which warrant further investigation for applications toward suppression and tolerance.


Assuntos
Células Dendríticas , Tolerância Imunológica , Animais , Camundongos , Análise em Microsséries
17.
Front Immunol ; 12: 653088, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122410

RESUMO

Allogeneic islet transplantation is a promising cell-based therapy for Type 1 Diabetes (T1D). The long-term efficacy of this approach, however, is impaired by allorejection. Current clinical practice relies on long-term systemic immunosuppression, leading to severe adverse events. To avoid these detrimental effects, poly(lactic-co-glycolic acid) (PLGA) microparticles (MPs) were engineered for the localized and controlled release of immunomodulatory TGF-ß1. The in vitro co-incubation of TGF-ß1 releasing PLGA MPs with naïve CD4+ T cells resulted in the efficient generation of both polyclonal and antigen-specific induced regulatory T cells (iTregs) with robust immunosuppressive function. The co-transplantation of TGF-ß1 releasing PLGA MPs and Balb/c mouse islets within the extrahepatic epididymal fat pad (EFP) of diabetic C57BL/6J mice resulted in the prompt engraftment of the allogenic implants, supporting the compatibility of PLGA MPs and local TGF-ß1 release. The presence of the TGF-ß1-PLGA MPs, however, did not confer significant graft protection when compared to untreated controls, despite measurement of preserved insulin expression, reduced intra-islet CD3+ cells invasion, and elevated CD3+Foxp3+ T cells at the peri-transplantation site in long-term functioning grafts. Examination of the broader impacts of TGF-ß1/PLGA MPs on the host immune system implicated a localized nature of the immunomodulation with no observed systemic impacts. In summary, this approach establishes the feasibility of a local and modular microparticle delivery system for the immunomodulation of an extrahepatic implant site. This approach can be easily adapted to deliver larger doses or other agents, as well as multi-drug approaches, within the local graft microenvironment to prevent transplant rejection.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Portadores de Fármacos/química , Rejeição de Enxerto/prevenção & controle , Fatores Imunológicos/administração & dosagem , Transplante das Ilhotas Pancreáticas/efeitos adversos , Fator de Crescimento Transformador beta1/administração & dosagem , Animais , Glicemia/análise , Técnicas de Cocultura , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Liberação Controlada de Fármacos , Estudos de Viabilidade , Teste de Tolerância a Glucose , Rejeição de Enxerto/imunologia , Humanos , Imunomodulação , Transplante das Ilhotas Pancreáticas/métodos , Masculino , Camundongos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Cultura Primária de Células , Ratos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Estreptozocina/administração & dosagem , Estreptozocina/toxicidade , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/farmacocinética , Transplante Homólogo/efeitos adversos
18.
iScience ; 24(4): 102307, 2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33870128

RESUMO

Mucosal-associated invariant T (MAIT) cells recognize microbial riboflavin metabolites presented by MR1 and play role in immune responses to microbial infections and tumors. We report here that absence of the transcription factor (TF) Bcl11b in mice alters predominantly MAIT17 cells in the thymus and further in the lung, both at steady state and following Salmonella infection. Transcriptomics and ChIP-seq analyses show direct control of TCR signaling program and position BCL11B upstream of essential TFs of MAIT17 program, including RORγt, ZBTB16 (PLZF), and MAF. BCL11B binding at key MAIT17 and at TCR signaling program genes in human MAIT cells occurred mostly in regions enriched for H3K27Ac. Unexpectedly, in human MAIT cells, BCL11B also bound at MAIT1 program genes, at putative active enhancers, although this program was not affected in mouse MAIT cells in the absence of Bcl11b. These studies endorse BCL11B as an essential TF for MAIT cells both in mice and humans.

19.
Adv Healthc Mater ; 9(11): e2000164, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32519501

RESUMO

Autoimmune diseases affect 10% of the world's population, and 1 in 200 people worldwide suffer from either multiple sclerosis (MS) or type 1 diabetes (T1D). While the targeted organ systems are different, MS and T1D share similarities in terms of autoreactive immune cells playing a critical role in pathogenesis. Both diseases can be managed only symptomatically without curative remission, and treatment options are limited and non-specific. Most current therapies cause some degree of systemic immune suppression, leaving the patients susceptible to opportunistic infections and other complications. Thus, there is considerable interest in the development of immunotherapies not associated with generalized immune suppression for these diseases. This review presents current and preclinical strategies for MS and T1D treatment, emphasizing those aimed to modulate the immune response, including the most recent strategies for tolerance induction. A central focus is on the emerging approaches using nano- and microparticle platforms, their evolution as immunotherapeutic carriers, including those incorporating specific antigens to induce tolerance and reduce unwanted generalized immune suppression.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1 , Esclerose Múltipla , Diabetes Mellitus Tipo 1/tratamento farmacológico , Humanos , Tolerância Imunológica , Imunoterapia , Esclerose Múltipla/tratamento farmacológico
20.
Front Immunol ; 11: 574447, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193362

RESUMO

Current monotherapeutic agents fail to restore tolerance to self-antigens in autoimmune individuals without systemic immunosuppression. We hypothesized that a combinatorial drug formulation delivered by a poly-lactic-co-glycolic acid (PLGA) dual-sized microparticle (dMP) system would facilitate tunable drug delivery to elicit immune tolerance. Specifically, we utilized 30 µm MPs to provide local sustained release of granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor ß1 (TGF-ß1) along with 1 µm MPs to facilitate phagocytic uptake of encapsulated antigen and 1α,25(OH)2 Vitamin D3 (VD3) followed by tolerogenic antigen presentation. We previously demonstrated the dMP system ameliorated type 1 diabetes (T1D) and experimental autoimmune encephalomyelitis (EAE) in murine models. Here, we investigated the system's capacity to impact human cell activity in vitro to advance clinical translation. dMP treatment directly reduced T cell proliferation and inflammatory cytokine production. dMP delivery to monocytes and monocyte-derived dendritic cells (DCs) increased their expression of surface and intracellular anti-inflammatory mediators. In co-culture, dMP-treated DCs (dMP-DCs) reduced allogeneic T cell receptor (TCR) signaling and proliferation, while increasing PD-1 expression, IL-10 production, and regulatory T cell (Treg) frequency. To model antigen-specific activation and downstream function, we co-cultured TCR-engineered autoreactive T cell "avatars," with dMP-DCs or control DCs followed by ß-cell line (ßlox5) target cells. For G6PC2-specific CD8+ avatars (clone 32), dMP-DC exposure reduced Granzyme B and dampened cytotoxicity. GAD65-reactive CD4+ avatars (clone 4.13) exhibited an anergic/exhausted phenotype with dMP-DC presence. Collectively, these data suggest this dMP formulation conditions human antigen presenting cells toward a tolerogenic phenotype, inducing regulatory and suppressive T cell responses.


Assuntos
Células Dendríticas/efeitos dos fármacos , Diabetes Mellitus Tipo 1/imunologia , Tolerância Imunológica/efeitos dos fármacos , Linfócitos T/imunologia , Apresentação de Antígeno/efeitos dos fármacos , Autoantígenos/imunologia , Calcitriol/química , Calcitriol/farmacologia , Células Dendríticas/imunologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/química , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Imunomodulação , Ativação Linfocitária , Monócitos/efeitos dos fármacos , Tamanho da Partícula , Fenótipo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Fator de Crescimento Transformador beta1/química , Fator de Crescimento Transformador beta1/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA