Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
2.
Sci Transl Med ; 14(654): eabn1413, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35857825

RESUMEN

To combat the HIV epidemic and emerging threats such as SARS-CoV-2, immunization strategies are needed that elicit protection at mucosal portals of pathogen entry. Immunization directly through airway surfaces is effective in driving mucosal immunity, but poor vaccine uptake across the mucus and epithelial lining is a limitation. The major blood protein albumin is constitutively transcytosed bidirectionally across the airway epithelium through interactions with neonatal Fc receptors (FcRn). Exploiting this biology, here, we demonstrate a strategy of "albumin hitchhiking" to promote mucosal immunity using an intranasal vaccine consisting of protein immunogens modified with an amphiphilic albumin-binding polymer-lipid tail, forming amph-proteins. Amph-proteins persisted in the nasal mucosa of mice and nonhuman primates and exhibited increased uptake into the tissue in an FcRn-dependent manner, leading to enhanced germinal center responses in nasal-associated lymphoid tissue. Intranasal immunization with amph-conjugated HIV Env gp120 or SARS-CoV-2 receptor binding domain (RBD) proteins elicited 100- to 1000-fold higher antigen-specific IgG and IgA titers in the serum, upper and lower respiratory mucosa, and distal genitourinary mucosae of mice compared to unmodified protein. Amph-RBD immunization induced high titers of SARS-CoV-2-neutralizing antibodies in serum, nasal washes, and bronchoalveolar lavage. Furthermore, intranasal amph-protein immunization in rhesus macaques elicited 10-fold higher antigen-specific IgG and IgA responses in the serum and nasal mucosa compared to unmodified protein, supporting the translational potential of this approach. These results suggest that using amph-protein vaccines to deliver antigen across mucosal epithelia is a promising strategy to promote mucosal immunity against HIV, SARS-CoV-2, and other infectious diseases.


Asunto(s)
COVID-19 , Infecciones por VIH , Administración Intranasal , Albúminas , Animales , Anticuerpos Antivirales , COVID-19/prevención & control , Infecciones por VIH/prevención & control , Inmunidad Mucosa , Inmunoglobulina A , Inmunoglobulina G , Lípidos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , SARS-CoV-2 , Vacunación
3.
Sci Immunol ; 6(66): eabf1152, 2021 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-34860581

RESUMEN

Saponins are potent and safe vaccine adjuvants, but their mechanisms of action remain incompletely understood. Here, we explored the properties of several saponin formulations, including immune-stimulatory complexes (ISCOMs) formed by the self-assembly of saponin and phospholipids in the absence or presence of the Toll-like receptor 4 agonist monophosphoryl lipid A (MPLA). We found that MPLA self-assembles with saponins to form particles physically resembling ISCOMs, which we termed saponin/MPLA nanoparticles (SMNP). Saponin-containing adjuvants exhibited distinctive mechanisms of action, altering lymph flow in a mast cell­dependent manner and promoting antigen entry into draining lymph nodes. SMNP was particularly effective, exhibiting even greater potency than the compositionally related adjuvant AS01B in mice, and primed robust germinal center B cell, TFH, and HIV tier 2 neutralizing antibodies in nonhuman primates. Together, these findings shed new light on mechanisms by which saponin adjuvants act to promote the immune response and suggest that SMNP may be a promising adjuvant in the setting of HIV, SARS-CoV-2, and other pathogens.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Linfa/efectos de los fármacos , Saponinas/farmacología , Receptores Toll-Like/agonistas , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Linfa/fisiología , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas , Ratas , Ratas Wistar
4.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34493582

RESUMEN

Global containment of COVID-19 still requires accessible and affordable vaccines for low- and middle-income countries (LMICs). Recently approved vaccines provide needed interventions, albeit at prices that may limit their global access. Subunit vaccines based on recombinant proteins are suited for large-volume microbial manufacturing to yield billions of doses annually, minimizing their manufacturing cost. These types of vaccines are well-established, proven interventions with multiple safe and efficacious commercial examples. Many vaccine candidates of this type for SARS-CoV-2 rely on sequences containing the receptor-binding domain (RBD), which mediates viral entry to cells via ACE2. Here we report an engineered sequence variant of RBD that exhibits high-yield manufacturability, high-affinity binding to ACE2, and enhanced immunogenicity after a single dose in mice compared to the Wuhan-Hu-1 variant used in current vaccines. Antibodies raised against the engineered protein exhibited heterotypic binding to the RBD from two recently reported SARS-CoV-2 variants of concern (501Y.V1/V2). Presentation of the engineered RBD on a designed virus-like particle (VLP) also reduced weight loss in hamsters upon viral challenge.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Ingeniería de Proteínas/métodos , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales , Sitios de Unión , COVID-19/virología , Vacunas contra la COVID-19/economía , Humanos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Unión Proteica , Conformación Proteica , Saccharomycetales/metabolismo , Vacunas de Subunidad
5.
Biomaterials ; 275: 120868, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34091299

RESUMEN

Antigen accumulation in lymph nodes (LNs) is critical for vaccine efficacy, but understanding of vaccine biodistribution in humans or large animals remains limited. Using the rhesus macaque model, we employed a combination of positron emission tomography (PET) and fluorescence imaging to characterize the whole-animal to tissue-level biodistribution of a subunit vaccine comprised of an HIV envelope trimer protein nanoparticle (trimer-NP) and lipid-conjugated CpG adjuvant (amph-CpG). Following immunization in the thigh, PET imaging revealed vaccine uptake primarily in inguinal and iliac LNs, reaching distances up to 17 cm away from the injection site. Within LNs, trimer-NPs exhibited striking accumulation on the periphery of follicular dendritic cell (FDC) networks in B cell follicles. Comparative imaging of soluble Env trimers (not presented on nanoparticles) in naïve or previously-immunized animals revealed diffuse deposition of trimer antigens in LNs following primary immunization, but concentration on FDCs in pre-immunized animals with high levels of trimer-specific IgG. These data demonstrate the capacity of nanoparticle or "albumin hitchhiking" technologies to concentrate vaccines in genitourinary tract-draining LNs, which may be valuable for promoting mucosal immunity.


Asunto(s)
Vacunas contra el SIDA , Vacunas , Adyuvantes Inmunológicos , Animales , Macaca mulatta , Tomografía de Emisión de Positrones , Distribución Tisular
6.
bioRxiv ; 2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33688647

RESUMEN

Global containment of COVID-19 still requires accessible and affordable vaccines for low- and middle-income countries (LMICs).1 Recently approved vaccines provide needed interventions, albeit at prices that may limit their global access.2 Subunit vaccines based on recombinant proteins are suited for large-volume microbial manufacturing to yield billions of doses annually, minimizing their manufacturing costs.3 These types of vaccines are well-established, proven interventions with multiple safe and efficacious commercial examples.4-6 Many vaccine candidates of this type for SARS-CoV-2 rely on sequences containing the receptor-binding domain (RBD), which mediates viral entry to cells via ACE2.7,8 Here we report an engineered sequence variant of RBD that exhibits high-yield manufacturability, high-affinity binding to ACE2, and enhanced immunogenicity after a single dose in mice compared to the Wuhan-Hu-1 variant used in current vaccines. Antibodies raised against the engineered protein exhibited heterotypic binding to the RBD from two recently reported SARS-CoV-2 variants of concern (501Y.V1/V2). Presentation of the engineered RBD on a designed virus-like particle (VLP) also reduced weight loss in hamsters upon viral challenge.

7.
Biomacromolecules ; 20(5): 2115-2122, 2019 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-30995843

RESUMEN

Antigen valency plays a fundamental role in directing the nature of an immune response to be stimulatory or tolerogenic. Soluble antigen arrays (SAgAs) are an antigen-specific immunotherapy that combats autoimmunity through the multivalent display of autoantigen. Although mechanistic studies have shown SAgAs to induce T- and B-cell anergy, the effect of SAgA valency has never been experimentally tested. Here, SAgAs of discrete antigen valencies were synthesized by click chemistry and evaluated for acute B-cell signaling inhibition as well as downstream immunomodulatory effects in splenocytes. Initial studies using the Raji B-cell line demonstrated SAgA valency dictated the extent of calcium flux. Lower valency constructs elicited the largest reductions in B-cell activation. In splenocytes from mice with experimental autoimmune encephalomyelitis, the same valency-dependent effects were evident in the downregulation of the costimulatory marker CD86. The reduction of calcium flux observed in Raji B-cells correlated strongly with downregulation in splenocyte CD86 expression after 72 h. Here, a thorough analysis of SAgA antigenic valency illustrates that low, but not monovalent, presentation of autoantigen was ideal for eliciting the most potent immunomodulatory effects.


Asunto(s)
Autoantígenos/química , Linfocitos B/inmunología , Inmunomodulación , Bazo/citología , Animales , Autoantígenos/inmunología , Antígeno B7-2/inmunología , Línea Celular Tumoral , Células Cultivadas , Química Clic/métodos , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Humanos , Inmunoensayo/métodos , Ratones , Análisis por Matrices de Proteínas/métodos , Bazo/inmunología
8.
J Autoimmun ; 93: 76-88, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30007842

RESUMEN

Autoreactive lymphocytes that escape central immune tolerance may be silenced via an endogenous peripheral tolerance mechanism known as anergy. Antigen-specific therapies capable of inducing anergy may restore patients with autoimmune diseases to a healthy phenotype while avoiding deleterious side effects associated with global immunosuppression. Inducing anergy in B cells may be a particularly potent intervention, as B cells can contribute to autoimmune diseases through multiple mechanisms and offer the potential for direct antigen-specific targeting through the B cell receptor (BCR). Our previous results suggested autoreactive B cells may be silenced by multivalent 'soluble antigen arrays' (SAgAs), which are polymer conjugates displaying multiple copies of autoantigen with or without a secondary peptide that blocks intracellular cell-adhesion molecule-1 (ICAM-1). Here, key therapeutic molecular properties of SAgAs were identified and linked to the immunological mechanism through comprehensive cellular and in vivo analyses. We determined non-hydrolyzable 'cSAgAs' displaying multivalent 'click'-conjugated antigen more potently suppressed experimental autoimmune encephalomyelitis (EAE) compared to hydrolyzable SAgAs capable of releasing conjugated antigen. cSAgAs restored a healthy phenotype in disease-specific antigen presenting cells (APCs) by inducing an anergic response in B cells and a subset of B cells called autoimmune-associated B cells (ABCs) that act as potent APCs in autoimmune disease. Accompanied by a cytokine response skewed towards a Th2/regulatory phenotype, this generated an environment of autoantigenic tolerance. By identifying key therapeutic molecular properties and an immunological mechanism that drives SAgA efficacy, this work guides the design of antigen-specific immunotherapies capable of inducing anergy.


Asunto(s)
Autoantígenos/genética , Subgrupos de Linfocitos B/efectos de los fármacos , Anergia Clonal/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/terapia , Inmunoconjugados/farmacología , Inmunoterapia/métodos , Fragmentos de Péptidos/farmacología , Animales , Autoantígenos/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Química Clic , Células Dendríticas/inmunología , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Hidrólisis , Inmunoconjugados/química , Inyecciones Subcutáneas , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Ratones , Proteína Proteolipídica de la Mielina/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/inmunología , Análisis por Matrices de Proteínas , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Bazo/inmunología , Bazo/patología , Células Th2/inmunología , Células Th2/patología
9.
J Control Release ; 266: 156-165, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-28963036

RESUMEN

Current therapies for autoimmune diseases focus on treating the symptoms rather than the underlying disease cause. A major setback in improving current therapeutics for autoimmunity is the lack of antigen specificity. Successful antigen-specific immunotherapy (ASIT) would allow for improved treatment of autoimmune diseases. In this work, dexamethasone was co-delivered with autoantigen (PLP) in vivo to create effective ASIT for the treatment of experimental autoimmune encephalomyelitis (EAE). Using an emulsion of incomplete Freund's adjuvant (IFA) as a co-delivery vehicle, it was discovered that the controlled release of autoantigen was important for the suppression of clinical disease symptoms. Analysis of the immune response via cytokines revealed that dexamethasone was important for shifting the immune response away from inflammation. Co-delivery of both autoantigen and dexamethasone increased B-cell populations and antibody production, signifying an increased humoral immune response. Overall, this data indicated that the co-delivery of PLP and dexamethasone with a water-in-oil emulsion is effective in treating a murine autoimmune model.


Asunto(s)
Antiinflamatorios/administración & dosificación , Autoantígenos/administración & dosificación , Dexametasona/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Adyuvante de Freund/administración & dosificación , Factores Inmunológicos/administración & dosificación , Lípidos/administración & dosificación , Proteína Proteolipídica de la Mielina/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Ratones , Bazo/citología
10.
Biomacromolecules ; 18(6): 1893-1907, 2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28474886

RESUMEN

A pressing need exists for antigen-specific immunotherapies (ASIT) that induce selective tolerance in autoimmune disease while avoiding deleterious global immunosuppression. Multivalent soluble antigen arrays (SAgAPLP:LABL), consisting of a hyaluronic acid (HA) linear polymer backbone cografted with multiple copies of autoantigen (PLP) and cell adhesion inhibitor (LABL) peptides, are designed to induce tolerance to a specific multiple sclerosis (MS) autoantigen. Previous studies established that hydrolyzable SAgAPLP:LABL, employing a degradable linker to codeliver PLP and LABL, was therapeutic in experimental autoimmune encephalomyelitis (EAE) in vivo and exhibited antigen-specific binding with B cells, targeted the B cell receptor (BCR), and dampened BCR-mediated signaling in vitro. Our results pointed to sustained BCR engagement as the SAgAPLP:LABL therapeutic mechanism, so we developed a new version of the SAgA molecule using nonhydrolyzable conjugation chemistry, hypothesizing it would enhance and maintain the molecule's action at the cell surface to improve efficacy. "Click SAgA" (cSAgAPLP:LABL) uses hydrolytically stable covalent conjugation chemistry (Copper-catalyzed Azide-Alkyne Cycloaddition (CuAAC)) rather than a hydrolyzable oxime bond to attach PLP and LABL to HA. We explored cSAgAPLP:LABL B cell engagement and modulation of BCR-mediated signaling in vitro through flow cytometry binding and calcium flux signaling assays. Indeed, cSAgAPLP:LABL exhibited higher avidity B cell binding and greater dampening of BCR-mediated signaling than hydrolyzable SAgAPLP:LABL. Furthermore, cSAgAPLP:LABL exhibited significantly enhanced in vivo efficacy compared to hydrolyzable SAgAPLP:LABL, achieving equivalent efficacy at one-quarter of the dose. These results indicate that nonhydrolyzable conjugation increased the avidity of cSAgAPLP:LABL to drive in vivo efficacy through modulated BCR-mediated signaling.


Asunto(s)
Linfocitos B/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/terapia , Inmunoconjugados/farmacología , Análisis por Matrices de Proteínas/métodos , Receptores de Antígenos de Linfocitos B/antagonistas & inhibidores , Alquinos/química , Animales , Autoantígenos/química , Autoantígenos/genética , Autoantígenos/inmunología , Azidas/química , Linfocitos B/inmunología , Linfocitos B/patología , Señalización del Calcio , Adhesión Celular/efectos de los fármacos , Línea Celular , Reacción de Cicloadición , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica , Humanos , Ácido Hialurónico/química , Tolerancia Inmunológica/efectos de los fármacos , Inmunoconjugados/química , Inmunoconjugados/genética , Inyecciones Subcutáneas , Ratones , Péptidos/síntesis química , Péptidos/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología
11.
Mol Pharm ; 14(1): 66-80, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28043135

RESUMEN

Current therapies to treat autoimmune diseases often result in side effects such as nonspecific immunosuppression. Therapies that can induce antigen-specific immune tolerance provide an opportunity to reverse autoimmunity and mitigate the risks associated with global immunosuppression. In an effort to induce antigen-specific immune tolerance, co-administration of immunomodulators with autoantigens has been investigated in an effort to reprogram autoimmunity. To date, identifying immunomodulators that may skew the antigen-specific immune response has been ad hoc at best. To address this need, we utilized splenocytes obtained from mice with experimental autoimmune encephalomyelitis (EAE) in order to determine if certain immunomodulators may induce markers of immune tolerance following antigen rechallenge. Of the immunomodulatory compounds investigated, only dexamethasone modified the antigen-specific immune response by skewing the cytokine response and decreasing T-cell populations at a concentration corresponding to a relevant in vivo dose. Thus, antigen-educated EAE splenocytes provide an ex vivo screen for investigating compounds capable of skewing the antigen-specific immune response, and this approach could be extrapolated to antigen-educated cells from other diseases or human tissues.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad/inmunología , Factores Inmunológicos/inmunología , Animales , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Tolerancia Inmunológica/inmunología , Ratones , Linfocitos T/inmunología
12.
Biomacromolecules ; 17(3): 710-22, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26771518

RESUMEN

A pressing need exists for autoimmune disease therapies that act in an antigen-specific manner while avoiding global immunosuppression. Multivalent soluble antigen arrays (SAgAPLP:LABL), designed to induce tolerance to a specific multiple sclerosis autoantigen, consist of a flexible hyaluronic acid (HA) polymer backbone cografted with multiple copies of autoantigen peptide (PLP) and cell adhesion inhibitor peptide (LABL). Previous in vivo studies revealed copresentation of both signals on HA was necessary for therapeutic efficacy. To elucidate therapeutic cellular mechanisms, in vitro studies were performed in a model B cell system to evaluate binding and specificity. Compared to HA and HA arrays containing only grafted PLP or LABL, SAgAPLP:LABL displaying both PLP and LABL exhibited greatly enhanced B cell binding. Furthermore, the binding avidity of SAgAPLP:LABL was primarily driven by the PLP antigen, determined via flow cytometry competitive dissociation studies. Fluorescence microscopy showed SAgAPLP:LABL induced mature receptor clustering that was faster than other HA arrays with only one type of grafted peptide. SAgAPLP:LABL molecules also reduced and inhibited IgM-stimulated signaling as discerned by a calcium flux assay. The molecular mechanisms of enhanced antigen-specific binding, mature receptor clustering, and dampened signaling observed in B cells may contribute to SAgAPLP:LABL therapeutic efficacy.


Asunto(s)
Autoantígenos/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Autoantígenos/química , Linfocitos B/inmunología , Línea Celular , Humanos , Ácido Hialurónico/química , Esclerosis Múltiple/inmunología , Análisis por Matrices de Proteínas , Transducción de Señal
13.
Mol Pharm ; 13(2): 330-43, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26636828

RESUMEN

Many current therapies for autoimmune diseases such as multiple sclerosis (MS) result in global immunosuppression, rendering insufficient efficacy with increased risk of adverse side effects. Multivalent soluble antigen arrays, nanomaterials presenting both autoantigen and secondary inhibitory signals on a flexible polymer backbone, are hypothesized to shift the immune response toward selective autoantigenic tolerance to repress autoimmune disease. Two-signal co-delivery of both autoantigen and secondary signal were deemed necessary for therapeutic efficacy against experimental autoimmune encephalomyelitis, a murine model of MS. Dynamic light scattering and in silico molecular dynamics simulations complemented these studies to illuminate the role of two-signal co-delivery in determining therapeutic potential. Physicochemical characteristics such as particle size and molecular affinity for intermolecular interactions and chain entanglement likely facilitated cotransport of two signals to produce efficacy. These findings elucidate potential mechanisms whereby soluble antigen arrays enact their therapeutic effect and help to guide the development of future multivalent antigen-specific immunotherapies.


Asunto(s)
Autoantígenos/inmunología , Sistemas de Liberación de Medicamentos , Encefalomielitis Autoinmune Experimental/terapia , Tolerancia Inmunológica/inmunología , Nanoestructuras/química , Polímeros/química , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Inmunoterapia , Ratones , Simulación de Dinámica Molecular , Análisis por Matrices de Proteínas
14.
J Pharm Sci ; 104(2): 346-61, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25447598

RESUMEN

Continued development of multivalent nanomaterials has provided opportunities for the advancement of antigen-specific immunotherapies. New insights emerge when considering the backdrop of vaccine design, which has long employed multivalent presentation of antigen to more strongly engage and enhance an immunogenic response. Additionally, vaccines traditionally codeliver antigen with adjuvant to amplify a robust antigen-specific response. Multivalent nanomaterials have since evolved for applications where immune tolerance is desired, such as autoimmune diseases or allergies. In particular, soluble, linear polymers may be tailored to direct antigen-specific immunogenicity or tolerance by modulating polymer length, ligand valency (number), and ligand density, in addition to incorporating secondary signals. Codelivery of a secondary signal may direct, amplify, or suppress the response to a given antigen. Although the ability of multivalent nanomaterials to enact an immune response through molecular mechanisms has been established, a transport mechanism for biodistribution must also be considered. Both mechanisms are influenced by ligand display and other physical properties of the nanomaterial. This review highlights multivalent ligand display on linear polymers, the complex interplay of physical parameters in multivalent design, and the ability to direct the immune response by molecular and transport mechanisms.


Asunto(s)
Antígenos/inmunología , Inmunoterapia , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Vacunas/química , Vacunas/inmunología , Humanos , Ligandos , Polímeros/química
15.
AAPS J ; 16(6): 1204-13, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25297853

RESUMEN

Autoimmune diseases such as multiple sclerosis (MS) are characterized by the breakdown of immune tolerance to autoantigens. Targeting surface receptors on immune cells offers a unique strategy for reprogramming immune responses in autoimmune diseases. The B7 signaling pathway was targeted using adaptations of soluble antigen array (SAgA) technology achieved by covalently linking B7-binding peptides and disease causing autoantigen (proteolipid peptide (PLP)) to hyaluronic acid (HA). We hypothesized that co-delivery of a B7-binding peptide and autoantigen would suppress experimental autoimmune encephalomyelitis (EAE), a murine model of MS. Three independent B7-targeted SAgAs were created containing peptides to either inhibit or potentially stimulate the B7 signaling pathway. Surprisingly, all SAgAs were found to suppress EAE disease symptoms. Altered cytokine expression was observed in primary splenocytes isolated from SAgA-treated mice, indicating that SAgAs with different B7-binding peptides may suppress EAE through different immunological mechanisms. This antigen-specific immunotherapy using SAgAs can successfully suppress EAE through co-delivery of autoantigen and peptides targeting with the B7 signaling pathway.


Asunto(s)
Autoantígenos/inmunología , Antígenos B7/inmunología , Portadores de Fármacos/química , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Proteína Proteolipídica de la Mielina/inmunología , Fragmentos de Péptidos/inmunología , Péptidos/uso terapéutico , Animales , Células Cultivadas , Citocinas/biosíntesis , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Ácido Hialurónico/química , Tolerancia Inmunológica/inmunología , Ratones Endogámicos , Péptidos/administración & dosificación , Péptidos/química , Solubilidad , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA