Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 42(7): 868-883, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35510552

RESUMEN

BACKGROUND: Arteriovenous fistulae (AVF) are the gold standard for vascular access for hemodialysis. Although the vein must thicken and dilate for successful hemodialysis, excessive wall thickness leads to stenosis causing AVF failure. Since TGF-ß (transforming growth factor-beta) regulates ECM (extracellular matrix) deposition and smooth muscle cell (SMC) proliferation-critical components of wall thickness-we hypothesized that disruption of TGF-ß signaling prevents excessive wall thickening during venous remodeling. METHODS: A mouse aortocaval fistula model was used. SB431542-an inhibitor of TGF-ß receptor I-was encapsulated in nanoparticles and applied to the AVF adventitia in C57BL/6J mice. Alternatively, AVFs were created in mice with conditional disruption of TGF-ß receptors in either SMCs or endothelial cells. Doppler ultrasound was performed serially to confirm patency and to measure vessel diameters. AVFs were harvested at predetermined time points for histological and immunofluorescence analyses. RESULTS: Inhibition of TGF-ß signaling with SB431542-containing nanoparticles significantly reduced p-Smad2-positive cells in the AVF wall during the early maturation phase (days 7-21) and was associated with decreased AVF wall thickness that showed both decreased collagen density and decreased SMC proliferation. SMC-specific TGF-ß signaling disruption decreased collagen density but not SMC proliferation or wall thickness. Endothelial cell-specific TGF-ß signaling disruption decreased both collagen density and SMC proliferation in the AVF wall and was associated with reduced wall thickness, increased outward remodeling, and improved AVF patency. CONCLUSIONS: Endothelial cell-targeted TGF-ß inhibition may be a translational strategy to improve AVF patency.


Asunto(s)
Fístula Arteriovenosa , Derivación Arteriovenosa Quirúrgica , Animales , Colágeno , Modelos Animales de Enfermedad , Células Endoteliales , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta , Factores de Crecimiento Transformadores , Remodelación Vascular/fisiología
2.
Nanomedicine ; 40: 102477, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34740868

RESUMEN

Magnetic resonance is a key imaging tool for the detection of prostate cancer; however, better tools focusing on cancer specificity are required to distinguish benign from cancerous regions. We found higher expression of claudin-3 (CLDN-3) and -4 (CLDN-4) in higher grade than lower-grade human prostate cancer biopsies (n = 174), leading to the design of functionalized nanoparticles (NPs) with a non-toxic truncated version of the natural ligand Clostridium perfringens enterotoxin (C-CPE) that has a strong binding affinity to Cldn-3 and Cldn-4 receptors. We developed a first-of-its-type, C-CPE-NP-based MRI detection tool in a prostate tumor-bearing mouse model. NPs with an average diameter of 152.9 ±â€¯15.7 nm (RS1) had a 2-fold enhancement of tumor specificity compared to larger (421.2 ±â€¯33.8 nm) NPs (RS4). There was a 1.8-fold (P < 0.01) and 1.6-fold (P < 0.01) upregulation of the tumor-to-liver signal intensities of C-RS1 and C-RS4 (functionalized NPs) compared to controls, respectively. Also, tumor specificity was 3.1-fold higher (P < 0.001) when comparing C-RS1 to C-RS4. This detection tool improved tumor localization of contrast-enhanced MRI, supporting potential clinical applicability.


Asunto(s)
Nanopartículas , Neoplasias de la Próstata , Animales , Enterotoxinas/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/metabolismo
4.
Nat Biomed Eng ; 5(9): 983-997, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34616050

RESUMEN

Oral formulations of insulin are typically designed to improve its intestinal absorption and increase its blood bioavailability. Here we show that polymerized ursodeoxycholic acid, selected from a panel of bile-acid polymers and formulated into nanoparticles for the oral delivery of insulin, restored blood-glucose levels in mice and pigs with established type 1 diabetes. The nanoparticles functioned as a protective insulin carrier and as a high-avidity bile-acid-receptor agonist, increased the intestinal absorption of insulin, polarized intestinal macrophages towards the M2 phenotype, and preferentially accumulated in the pancreas of the mice, binding to the islet-cell bile-acid membrane receptor TGR5 with high avidity and activating the secretion of glucagon-like peptide and of endogenous insulin. In the mice, the nanoparticles also reversed inflammation, restored metabolic functions and extended animal survival. When encapsulating rapamycin, they delayed the onset of diabetes in mice with chemically induced pancreatic inflammation. The metabolic and immunomodulatory functions of ingestible bile-acid-polymer nanocarriers may offer translational opportunities for the prevention and treatment of type 1 diabetes.


Asunto(s)
Ácidos y Sales Biliares , Diabetes Mellitus Tipo 1 , Animales , Bilis , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Péptido 1 Similar al Glucagón , Insulina , Ratones , Polímeros , Receptores Acoplados a Proteínas G , Sirolimus , Porcinos
5.
J Colloid Interface Sci ; 587: 101-109, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33360882

RESUMEN

The unique capability of fullerene (C60) to absorb light and generate reactive oxygen species (ROS) has been extensively studied for photosensitized water treatment and cancer therapy. Various material synthesis strategies have been proposed in parallel to overcome its intrinsic hydrophobicity and to enhance availability in water and physiological media. We present here a strikingly simple approach to make C60 available to these applications by hand-grinding dry C60 powder with nanodiamond (ND) using a mortar and pestle. The resulting ND-C60 composite was found to form a stable aqueous colloidal suspension and efficiently drive photosensitized production of ROS under visible light illumination. ND-C60 rapidly adsorbed and oxidized organic contaminants by photogenerated ROS. In the experiments for photodynamic cancer therapy, ND-C60 was internalized by cancer cells and induced cell apoptosis without noticeable toxicity. Treatment of tumor-bearing mice with ND-C60 and light irradiation resulted in tumor shrinkage and prolonged survival time.


Asunto(s)
Fulerenos , Nanodiamantes , Neoplasias , Fotoquimioterapia , Purificación del Agua , Animales , Ratones , Neoplasias/tratamiento farmacológico
6.
ACS Nano ; 14(7): 8646-8657, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32530598

RESUMEN

The sensitivity and speed with which the immune system reacts to host disruption is unrivaled by any detection method for pathogenic biomarkers or infectious signatures. Engagement of cellular immunity in response to infections or cancer is contingent upon activation and subsequent cytotoxic activity by T cells. Thus, monitoring T cell activation can reliably serve as a metric for disease diagnosis as well as therapeutic prognosis. Rapid and direct quantification of T cell activation states, however, has been hindered by challenges associated with antigen target identification, labeling requirements, and assay duration. Here we present an electronic, label-free method for simultaneous separation and evaluation of T cell activation states. Our device utilizes a microfluidic design integrated with nanolayered electrode structures for dielectrophoresis (DEP)-driven discrimination of activated vs naïve T cells at single-cell resolution and demonstrates rapid (<2 min) separation of T cells at high single-pass efficiency as quantified by an on-chip Coulter counter module. Our device represents a microfluidic tool for electronic assessment of immune activation states and, hence, a portable diagnostic for quantitative evaluation of immunity and disease state. Further, its ability to achieve label-free enrichment of activated immune cells promises clinical utility in cell-based immunotherapies.


Asunto(s)
Microfluídica , Linfocitos T , Bioensayo , Separación Celular , Electrónica , Electroforesis , Activación de Linfocitos
7.
Front Immunol ; 11: 583338, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33391260

RESUMEN

We recently reported that the treatment with nanoparticles (NPs) loaded with tolerogenic cytokines suppressed the manifestations of lupus-like disease induced by the transfer of donor CD4+ T cells from DBA/2 mice into (C57BL/6 × DBA/2)F1 (BDF1) mice. Although the protective effects were ascribed to the induction of adaptive CD4+ and CD8+ T regulatory cells, the results suggested that another population of immune cells could be involved. Here we report that NK cells critically contribute to the protection from lupus-like disease conferred by NPs to BDF1 mice, and that this effect is TGF-ß-dependent.


Asunto(s)
Antígenos CD2/antagonistas & inhibidores , Interleucina-2/farmacología , Células Asesinas Naturales/inmunología , Lupus Eritematoso Sistémico/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Células Asesinas Naturales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Nanopartículas
8.
Trends Immunol ; 40(10): 888-908, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601519

RESUMEN

During homeostasis, interactions between tolerogenic dendritic cells (DCs), self-reactive T cells, and T regulatory cells (Tregs) contribute to maintaining mammalian immune tolerance. In response to infection, immunogenic DCs promote the generation of proinflammatory effector T cell subsets. When complex homeostatic mechanisms maintaining the balance between regulatory and effector functions become impaired, autoimmune diseases can develop. We discuss some of the newest advances on the mechanisms of physiopathologic homeostasis that can be employed to develop strategies to restore a dysregulated immune equilibrium. Some of these designs are based on selectively activating regulators of immunity and inflammation instead of broadly suppressing these processes. Promising approaches include the use of nanoparticles (NPs) to restore Treg control over self-reactive cells, aiming to achieve long-term disease remission, and potentially to prevent autoimmunity in susceptible individuals.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Homeostasis/inmunología , Animales , Autoinmunidad/inmunología , Células Dendríticas/inmunología , Humanos , Inflamación/inmunología , Linfocitos T/inmunología
9.
Arthritis Rheumatol ; 71(4): 632-640, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30407752

RESUMEN

OBJECTIVE: To develop a nanoparticle (NP) platform that can expand both CD4+ and CD8+ Treg cells in vivo for the suppression of autoimmune responses in systemic lupus erythematosus (SLE). METHODS: Poly(lactic-co-glycolic acid) (PLGA) NPs encapsulating interleukin-2 (IL-2) and transforming growth factor ß (TGFß) were coated with anti-CD2/CD4 antibodies and administered to mice with lupus-like disease induced by the transfer of DBA/2 T cells into (C57BL/6 × DBA/2)F1 (BDF1) mice. The peripheral frequency of Treg cells was monitored ex vivo by flow cytometry. Disease progression was assessed by measuring serum anti-double-stranded DNA antibody levels by enzyme-linked immunosorbent assay. Kidney disease was defined as the presence of proteinuria or renal histopathologic features. RESULTS: Anti-CD2/CD4 antibody-coated, but not noncoated, NPs encapsulating IL-2 and TGFß induced CD4+ and CD8+ FoxP3+ Treg cells in vitro. The optimal dosing regimen of NPs for expansion of CD4+ and CD8+ Treg cells was determined in in vivo studies in mice without lupus and then tested in BDF1 mice with lupus. The administration of anti-CD2/CD4 antibody-coated NPs encapsulating IL-2 and TGFß resulted in the expansion of CD4+ and CD8+ Treg cells, a marked suppression of anti-DNA antibody production, and reduced renal disease. CONCLUSION: This study shows for the first time that T cell-targeted PLGA NPs encapsulating IL-2 and TGFß can expand both CD4+ and CD8+ Treg cells in vivo and suppress murine lupus. This approach, which enables the expansion of Treg cells in vivo and inhibits pathogenic immune responses in SLE, could represent a potential new therapeutic modality in autoimmune conditions characterized by impaired Treg cell function associated with IL-2 deficiency.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Interleucina-2/administración & dosificación , Lupus Eritematoso Sistémico/tratamiento farmacológico , Nanopartículas/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos C57BL
10.
JCI Insight ; 3(16)2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30135300

RESUMEN

Defective DNA methylation in T cells leads to a series of T cell abnormalities in lupus; however, the full effect of T cell lineage-specific DNA methylation on disease expression has not been explored. Here, we show that 5-azacytidine, a DNA methyltransferase inhibitor, targeted to either CD4 or CD8 T cells in mice with established disease using a nanolipogel delivery system dramatically ameliorates lupus-related pathology through distinct mechanisms. In vivo targeted delivery of 5-azacytidine into CD4 T cells favors the expansion and function of Foxp3+ Tregs, whereas targeted delivery to CD8 T cells enhances the cytotoxicity and restrains the expansion of pathogenic TCR-αß+CD4-CD8- double-negative T cells. Our results signify the importance of cell-specific inhibition of DNA methylation in the treatment of established lupus.


Asunto(s)
Azacitidina/administración & dosificación , Metilación de ADN/efectos de los fármacos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Nanoconjugados/química , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Metilación de ADN/inmunología , Metilasas de Modificación del ADN/antagonistas & inhibidores , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/inmunología , Femenino , Humanos , Inmunoconjugados/química , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Transgénicos , Resultado del Tratamiento
11.
J Clin Invest ; 128(8): 3445-3459, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29985166

RESUMEN

Podocyte malfunction occurs in autoimmune and nonautoimmune kidney disease. Calcium signaling is essential for podocyte injury, but the role of Ca2+/calmodulin-dependent kinase (CaMK) signaling in podocytes has not been fully explored. We report that podocytes from patients with lupus nephritis and focal segmental glomerulosclerosis and lupus-prone and lipopolysaccharide- or adriamycin-treated mice display increased expression of CaMK IV (CaMK4), but not CaMK2. Mechanistically, CaMK4 modulated podocyte motility by altering the expression of the GTPases Rac1 and RhoA and suppressed the expression of nephrin, synaptopodin, and actin fibers in podocytes. In addition, it phosphorylated the scaffold protein 14-3-3ß, which resulted in the release and degradation of synaptopodin. Targeted delivery of a CaMK4 inhibitor to podocytes preserved their ultrastructure, averted immune complex deposition and crescent formation, and suppressed proteinuria in lupus-prone mice and proteinuria in mice exposed to lipopolysaccharide-induced podocyte injury by preserving nephrin/synaptopodin expression. In animals exposed to adriamycin, podocyte-specific delivery of a CaMK4 inhibitor prevented and reversed podocyte injury and renal disease. We conclude that CaMK4 is pivotal in immune and nonimmune podocyte injury and that its targeted cell-specific inhibition preserves podocyte structure and function and should have therapeutic value in lupus nephritis and podocytopathies, including focal segmental glomerulosclerosis.


Asunto(s)
Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Glomeruloesclerosis Focal y Segmentaria/enzimología , Glomérulos Renales/enzimología , Nefritis Lúpica/enzimología , Transducción de Señal , Animales , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/inmunología , Línea Celular Transformada , Femenino , Glomeruloesclerosis Focal y Segmentaria/inmunología , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Ratones Noqueados , Proteinuria/enzimología , Proteinuria/inmunología , Proteinuria/patología
12.
Arterioscler Thromb Vasc Biol ; 38(1): 195-205, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29146747

RESUMEN

OBJECTIVE: Pseudoaneurysms remain a significant complication after vascular procedures. We hypothesized that TGF-ß (transforming growth factor-ß) signaling plays a mechanistic role in the development of pseudoaneurysms. APPROACH AND RESULTS: Rat aortic pericardial patch angioplasty was associated with a high incidence (88%) of pseudoaneurysms at 30 days, with increased smad2 phosphorylation in small pseudoaneurysms but not in large pseudoaneurysms; TGF-ß1 receptors were increased in small pseudoaneurysms and preserved in large pseudoaneurysms. Delivery of TGF-ß1 via nanoparticles covalently bonded to the patch stimulated smad2 phosphorylation both in vitro and in vivo and significantly decreased pseudoaneurysm formation (6.7%). Inhibition of TGF-ß1 signaling with SB431542 decreased smad2 phosphorylation both in vitro and in vivo and significantly induced pseudoaneurysm formation by day 7 (66.7%). CONCLUSIONS: Normal healing after aortic patch angioplasty is associated with increased TGF-ß1 signaling, and recruitment of smad2 signaling may limit pseudoaneurysm formation; loss of TGF-ß1 signaling is associated with the formation of large pseudoaneurysms. Enhancement of TGF-ß1 signaling may be a potential mechanism to limit pseudoaneurysm formation after vascular intervention.


Asunto(s)
Aneurisma Falso/prevención & control , Angioplastia/instrumentación , Aorta/cirugía , Aneurisma de la Aorta/prevención & control , Materiales Biocompatibles Revestidos , Pericardio/trasplante , Factor de Crecimiento Transformador beta1/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Aneurisma Falso/etiología , Aneurisma Falso/metabolismo , Aneurisma Falso/patología , Angioplastia/efectos adversos , Animales , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta/etiología , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Células Cultivadas , Masculino , Ratones , Nanopartículas , Fosforilación , Diseño de Prótesis , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Factores de Tiempo
13.
Methods Mol Biol ; 1530: 343-353, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28150213

RESUMEN

Artificial antigen-presenting cells (aAPCs) overcome many of the limitations of biologically based adoptive immunotherapy protocols. While these acellular systems can be designed with a variety of parameters, including material type, diameter, and proliferative signals for T cells, we outline methods to formulate and characterize a comprehensive polymeric microparticle aAPC platform. These aAPCs, which can be reproducibly fabricated in large quantities, efficiently stimulate antigen-specific T cell activation and proliferation by both paracrine cytokine signals and engagement of T cell surface proteins.


Asunto(s)
Presentación de Antígeno , Células Artificiales/inmunología , Inmunoterapia , Anticuerpos/química , Anticuerpos/inmunología , Células Presentadoras de Antígenos/inmunología , Avidina , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/inmunología , Humanos , Inmunoterapia Adoptiva , Ácido Láctico/química , Microesferas , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Propiedades de Superficie , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
14.
Sci Rep ; 7: 40142, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28071663

RESUMEN

Prosthetic grafts and patches are commonly used in cardiovascular surgery, however neointimal hyperplasia remains a significant concern, especially under low flow conditions. We hypothesized that delivery of rapamycin from nanoparticles (NP) covalently attached to patches allows sustained site-specific delivery of therapeutic agents targeted to inhibit localized neointimal hyperplasia. NP were covalently linked to pericardial patches using EDC/NHS chemistry and could deliver at least 360 ng rapamycin per patch without detectable rapamycin in serum; nanoparticles were detectable in the liver, kidney and spleen but no other sites within 24 hours. In a rat venous patch angioplasty model, control patches developed robust neointimal hyperplasia on the patch luminal surface characterized by Eph-B4-positive endothelium and underlying SMC and infiltrating cells such as macrophages and leukocytes. Patches delivering rapamycin developed less neointimal hyperplasia, less smooth muscle cell proliferation, and had fewer infiltrating cells but retained endothelialization. NP covalently linked to pericardial patches are a novel composite delivery system that allows sustained site-specific delivery of therapeutics; NP delivering rapamycin inhibit patch neointimal hyperplasia. NP linked to patches may represent a next generation of tissue engineered cardiovascular implants.


Asunto(s)
Angioplastia/métodos , Inhibidores de Crecimiento/administración & dosificación , Hiperplasia/prevención & control , Nanopartículas/administración & dosificación , Neointima/patología , Sirolimus/administración & dosificación , Trasplantes/cirugía , Animales , Portadores de Fármacos/administración & dosificación , Inhibidores de Crecimiento/farmacocinética , Histocitoquímica , Hiperplasia/patología , Ratas , Sirolimus/farmacocinética , Resultado del Tratamiento
15.
Acta Biomater ; 57: 136-145, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069499

RESUMEN

The biophysical parameters governing nanoparticle (NP)-cell interactions significantly affect biological responses, particularly in the application of NP-based immunotherapeutics. Modulation of the surface biophysical character of NPs can be achieved via introduction of amino acids, which offer the ability to fine tune a range of biophysical parameters of interest. We employed this approach using monodisperse silica NPs coated with numerous poly(amino acid)s (PAAs). The NPs were incubated with dendritic cells (DCs) in conjunction with TLR ligands and production of IL-1ß from DCs and IFNγ from T cells primed by these DCs were measured. These key cytokines can prognosticate the efficacy of the NP platform as a potential vaccine or active cellular immunotherapy carrier. IL-1ß production showed a correlation with both NP size and degree of hydrophobicity. High IFNγ secretion from T cells was shown to be correlated with both the hydrophobicity and charge of the NPs used to activate the DCs. Other cytokines were also screened in order to compare the immune responses. The results of this study highlight the importance of nanoparticle biophysical parameters and the selection of TLR ligands to the rational design of nanoparticle-based vaccines and immunotherapies. STATEMENT OF SIGNIFICANCE: The manuscript describes a systematic investigation into the effects of biophysical parameters of nanoparticles (NPs) on immune cells. Modulation of the biophysical character of the NP surface can be achieved by introduction of amino acids on monodisperse silica NPs, introducing a range of tunable biophysical parameters of interest, i.e. distinct sizes, different surface charges and varying degrees of surface hydrophobicity. We examine internalization of the NP in dendritic cells (DCs) and measure a myriad of cytokines, including IL-1ß and IFNγ, which prognosticate the efficacy of the NPs as a potential vaccine (IL-1ß metric) or active cellular immunotherapy carrier (IFNγ metric). Two different TLR ligands (a viral TLR3 ligand and a bacterial TLR4 ligand) were used along with the PAA NPs to compare their costimulatory immunogenicity. We strongly believe that this study will provide crucial information to many readers of Acta Biomaterialia and further drive the use of nanoparticle platforms in modulating immune responses.


Asunto(s)
Células Dendríticas/inmunología , Interferón gamma/inmunología , Interleucina-1beta/inmunología , Nanopartículas/química , Linfocitos T/inmunología , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 4/agonistas , Animales , Femenino , Lignanos , Ratones , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 4/inmunología
16.
Biomaterials ; 108: 168-76, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27636154

RESUMEN

Leishmania (Viannia) panamensis (L. (V.) panamensis) is a species of protozoan parasites that causes New World leishmaniasis, which is characterized by a hyper-inflammatory response. Current treatment strategies, mainly chemotherapeutic, are suboptimal due to adverse effects, long treatment regimens, and increasing drug resistance. Recently, immunotherapeutic approaches have shown promise in preclinical studies of leishmaniasis. As NPs may enable broad cellular immunomodulation through internalization in phagocytic and antigen-presenting cells, we tested the therapeutic efficacy of biodegradable NPs encapsulating a pathogen-associated molecular pattern (PAMP), CpG-rich oligonucleotide (CpG; NP-CpG), in mice infected with L. (V.) panamensis. NP-CpG treatment reduced lesion size and parasite burden, while neither free CpG nor empty NP showed therapeutic effects. NP-encapsulation led to CpG persistence at the site of infection along with an unexpected preferential cellular uptake by myeloid derived suppressor cells (MDSCs; CD11b(+)Ly6G(+)Ly6C(-)) as well as CD19(+) dendritic cells. This corresponded with the suppression of the ongoing immune response measured by the reduction of pathogenic cytokines IL-10 and IL-13, as well as IL-17 and IFNγ, in comparison to other treatment groups. As chronic inflammation is generally associated with the accumulation of MDSCs, this study may enable the rational design of cost-effective, safe, and scalable delivery systems for the treatment of inflammation-mediated diseases.


Asunto(s)
Citocinas/inmunología , Preparaciones de Acción Retardada/administración & dosificación , Factores Inmunológicos/administración & dosificación , Leishmaniasis/inmunología , Leishmaniasis/terapia , Nanopartículas/administración & dosificación , Animales , Preparaciones de Acción Retardada/química , Femenino , Factores Inmunológicos/química , Leishmania , Leishmaniasis/parasitología , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/ultraestructura , Resultado del Tratamiento
17.
J Allergy Clin Immunol ; 138(2): 536-543.e4, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27130858

RESUMEN

BACKGROUND: Treatments to reverse peanut allergy remain elusive. Current clinical approaches using peanut oral/sublingual immunotherapy are promising, but concerns about safety and long-term benefit remain a barrier to wide use. Improved methods of delivering peanut-specific immunotherapy are needed. OBJECTIVE: We sought to investigate the efficacy and safety of peanut oral immunotherapy using CpG-coated poly(lactic-co-glycolic acid) nanoparticles containing peanut extract (CpG/PN-NPs) in a murine model of peanut allergy. METHODS: C3H/HeJ mice were rendered peanut allergic by means of oral sensitization with peanut and cholera toxin. Mice were then subjected to 4 weekly gavages with CpG/PN-NPs, vehicle (PBS), nanoparticles alone, peanut alone, CpG nanoparticles, or peanut nanoparticles. Untreated mice served as naive controls. After completing therapy, mice underwent 5 monthly oral peanut challenges. Anaphylaxis was evaluated by means of visual assessment of symptom scores and measurement of body temperature and plasma histamine levels. Peanut-specific serum IgE, IgG1, and IgG2a levels were measured by using ELISA, as were cytokine recall responses in splenocyte cultures. RESULTS: Mice with peanut allergy treated with CpG/PN-NPs but not vehicle or other treatment components were significantly protected from anaphylaxis to all 5 oral peanut challenges, as indicated by lower symptom scores, less change in body temperature, and a lower increase of plasma histamine levels. Importantly, CpG/PN-NP treatment did not cause anaphylactic reactions. Treatment was associated with a sustained and significant decrease in peanut-specific IgE/IgG1 levels and an increase in peanut-specific IgG2a levels. Compared with vehicle control animals, peanut recall responses in splenocyte cultures from nanoparticle-treated mice showed significantly decreased levels of TH2 cytokines (IL-4, IL-5, and IL-13) but increased IFN-γ levels in cell supernatants. CONCLUSIONS: Preclinical findings indicate that peanut oral immunotherapy with CpG/PN-NPs might be a valuable strategy for peanut-specific immunotherapy in human subjects.


Asunto(s)
Alérgenos/inmunología , Arachis/efectos adversos , Desensibilización Inmunológica , Ácido Láctico , Nanopartículas , Hipersensibilidad al Cacahuete/inmunología , Ácido Poliglicólico , Alérgenos/administración & dosificación , Animales , Citocinas/sangre , Citocinas/metabolismo , Desensibilización Inmunológica/métodos , Modelos Animales de Enfermedad , Femenino , Histamina/sangre , Inmunización , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Hipersensibilidad al Cacahuete/diagnóstico , Hipersensibilidad al Cacahuete/metabolismo , Hipersensibilidad al Cacahuete/terapia , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Extractos Vegetales/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
18.
Biomaterials ; 97: 85-96, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27162077

RESUMEN

Antigen-presenting cells (APCs) sense microorganisms via pathogen-associated molecular patterns (PAMPs) by both extra- and intracellular Toll-like Receptors (TLRs), initiating immune responses against invading pathogens. Bacterial PAMPs include extracellular lipopolysaccharides and intracellular unmethylated CpG-rich oligodeoxynucleotides (CpG). We hypothesized that a biomimetic approach involving antigen-loaded nanoparticles (NP) displaying Monophosphoryl Lipid A (MPLA) and encapsulating CpG may function as an effective "artificial bacterial" biomimetic vaccine platform. This hypothesis was tested in vitro and in vivo using NP assembled from biodegradable poly(lactic-co-glycolic acid) (PLGA) polymer, surface-modified with MPLA, and loaded with CpG and model antigen Ovalbumin (OVA). First, CpG potency, characterized by cytokine profiles, titers, and antigen-specific T cell responses, was enhanced when CpG was encapsulated in NP compared to equivalent concentrations of surface-presented CpG, highlighting the importance of biomimetic presentation of PAMPs. Second, NP synergized surface-bound MPLA with encapsulated CpG in vitro and in vivo, inducing greater pro-inflammatory, antigen-specific T helper 1 (Th1)-skewed cellular and antibody-mediated responses compared to single PAMPs or soluble PAMP combinations. Importantly, NP co-presentation of CpG and MPLA was critical for CD8(+) T cell responses, as vaccination with a mixture of NP presenting either CpG or MPLA failed to induce cellular immunity. This work demonstrates a rational methodology for combining TLR ligands in a context-dependent manner for synergistic nanoparticulate vaccines.


Asunto(s)
Vacunas Bacterianas/inmunología , Materiales Biomiméticos/farmacología , Nanopartículas/química , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Animales , Antibacterianos/farmacología , Formación de Anticuerpos/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Humanos , Inmunidad Celular/efectos de los fármacos , Lípido A/análogos & derivados , Lípido A/química , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Receptores Toll-Like/metabolismo
19.
J Biol Chem ; 291(17): 8931-50, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26719339

RESUMEN

Here we report a "configuration-dependent" mechanism of action for IL-15:IL-15Rα (heterodimeric IL-15 or hetIL-15) where the manner by which IL-15:IL-15Rα molecules are presented to target cells significantly affects its function as a vaccine adjuvant. Although the cellular mechanism of IL-15 trans-presentation via IL-15Rα and its importance for IL-15 function have been described, the full effect of the IL-15:IL-15Rα configuration on responding cells is not yet known. We found that trans-presenting IL-15:IL-15Rα in a multivalent fashion on the surface of antigen-encapsulating nanoparticles enhanced the ability of nanoparticle-treated dendritic cells (DCs) to stimulate antigen-specific CD8(+) T cell responses. Localization of multivalent IL-15:IL-15Rα and encapsulated antigen to the same DC led to maximal T cell responses. Strikingly, DCs incubated with IL-15:IL-15Rα-coated nanoparticles displayed higher levels of functional IL-15 on the cell surface, implicating a mechanism for nanoparticle-mediated transfer of IL-15 to the DC surface. Using artificial antigen-presenting cells to highlight the effect of IL-15 configuration on DCs, we showed that artificial antigen-presenting cells presenting IL-15:IL-15Rα increased the sensitivity and magnitude of the T cell response, whereas IL-2 enhanced the T cell response only when delivered in a paracrine fashion. Therefore, the mode of cytokine presentation (configuration) is important for optimal immune responses. We tested the effect of configuration dependence in an aggressive model of murine melanoma and demonstrated significantly delayed tumor progression induced by IL-15:IL-15Rα-coated nanoparticles in comparison with monovalent IL-15:IL-15Rα. The novel mechanism of IL-15 transfer to the surface of antigen-processing DCs may explain the enhanced potency of IL-15:IL-15Rα-coated nanoparticles for antigen delivery.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Antígenos de Neoplasias , Linfocitos T CD8-positivos/inmunología , Materiales Biocompatibles Revestidos/farmacología , Células Dendríticas/inmunología , Inmunidad Celular/efectos de los fármacos , Interleucina-15 , Nanopartículas , Neoplasias Experimentales , Receptores de Interleucina-15/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Humanos , Interleucina-15/inmunología , Interleucina-15/farmacología , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología
20.
J Immunol ; 195(12): 5533-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26561550

RESUMEN

Treatment of autoimmune diseases is still largely based on the use of systemically acting immunosuppressive drugs, which invariably cause severe side effects. Calcium/calmodulin-dependent protein kinase IV is involved in the suppression of IL-2 and the production of IL-17. Its pharmacologic or genetic inhibition limits autoimmune disease in mice. In this study, we demonstrate that KN93, a small-molecule inhibitor of calcium/calmodulin-dependent protein kinase IV, targeted to CD4(+) T cells via a nanolipogel delivery system, markedly reduced experimental autoimmune encephalomyelitis and was 10-fold more potent than the free systemically delivered drug in the lupus mouse models. The targeted delivery of KN93 did not deplete T cells but effectively blocked Th17 cell differentiation and expansion as measured in the spinal cords and kidneys of mice developing experimental autoimmune encephalomyelitis or lupus, respectively. These results highlight the promise of cell-targeted inhibition of molecules involved in the pathogenesis of autoimmunity as a means of advancing the treatment of autoimmune diseases.


Asunto(s)
Bencilaminas/administración & dosificación , Linfocitos T CD4-Positivos/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Polietileneimina/administración & dosificación , Sulfonamidas/administración & dosificación , Células Th17/efectos de los fármacos , Animales , Bencilaminas/farmacología , Linfocitos T CD4-Positivos/inmunología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Terapia de Inmunosupresión , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Nanogeles , Sulfonamidas/farmacología , Células Th17/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA