Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Toxicol Appl Pharmacol ; 434: 115796, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34785274

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has required the urgent development of new therapies, among which passive immunotherapy is contemplated. CoviFab (INM005) is a RBD-specific F(ab')2 fragment derived from equine polyclonal antibodies. We investigate their preclinical security and biodistribution by in vivo and ex vivo NIR imaging after intravenous administration of a dose of 4 mg/kg at time 0 and 48 h. Images were taken at 1, 12, 24, 36, 48, 49, 60, 72, 84, 96, 108, 120, 132 and 144 h after the first intravenous injection. At 96 and 144 h, mice were sacrificed for haematology, serum chemistry, clinical pathology, histopathology and ex vivo imaging. The biodistribution profile was similar in all organs studied, with the highest fluorescence at 1 h after each injection, gradually decreasing after that each one and until the end of the study (144 h). The toxicology study revealed no significant changes in the haematology and serum chemistry parameters. Further, there were no changes in the gross and histological examination of organs. Nonclinical data of the current study confirm that CoviFab is safe, without observable adverse effects in mice. Furthermore, we confirm that bioimaging studies are a useful approach in preclinical trials to determine biodistribution.


Asunto(s)
Anticuerpos Antivirales/metabolismo , Tratamiento Farmacológico de COVID-19 , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/metabolismo , SARS-CoV-2/metabolismo , Administración Intravenosa , Animales , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/efectos adversos , COVID-19/metabolismo , COVID-19/prevención & control , Células HEK293 , Caballos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptores Inmunológicos/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , SARS-CoV-2/efectos de los fármacos , Espectroscopía Infrarroja Corta/métodos , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
2.
Cancer Gene Ther ; 28(9): 960-970, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34349240

RESUMEN

Antigen-presenting cells (APCs), including macrophages and dendritic cells (DCs), play a crucial role in bridging innate and adaptive immunity; thereby, innate immune checkpoint blockade-based therapy is an attractive approach for the induction of sustainable tumor-specific immunity. The interaction between the cluster of differentiation 47 (CD47) on tumor and signal-regulatory protein alpha (SIRPα) on phagocytic cells inhibits the phagocytic function of APCs, acting as a "don't eat me" signal. Accordingly, CD47 blockade is known to increase tumor cell phagocytosis, eliciting tumor-specific CD8+ T-cell immunity. Here, we introduced a nature-derived nanocage to deliver SIRPγ for blocking of antiphagocytic signaling through binding to CD47 and combined it with prophagocytic stimuli using a metabolic reprogramming reagent for APCs (CpG-oligodeoxynucleotides). Upon delivering the clustered SIRPγ variant, the nanocage showed enhanced CD47 binding profiles on tumor cells, thereby promoting active engulfment by phagocytes. Moreover, combination with CpG potentiated the prophagocytic ability, leading to the establishment of antitumorigenic surroundings. This combination treatment could competently inhibit tumor growth by invigorating APCs and CD8+ T-cells in TMEs in B16F10 orthotopic tumor models, known to be resistant to CD47-targeting therapeutics. Collectively, enhanced delivery of an innate immune checkpoint antagonist with metabolic modulation stimuli of immune cells could be a promising strategy for arousing immune responses against cancer.


Asunto(s)
Antígenos de Diferenciación/administración & dosificación , Antígenos de Diferenciación/inmunología , Ferritinas/administración & dosificación , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Nanoestructuras/uso terapéutico , Oxidorreductasas/administración & dosificación , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/inmunología , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ferritinas/química , Ferritinas/genética , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Nanoestructuras/química , Células Neoplásicas Circulantes/inmunología , Oxidorreductasas/química , Oxidorreductasas/genética , Fagocitosis/inmunología , Receptores Inmunológicos/química , Receptores Inmunológicos/genética
3.
Int Immunol ; 32(3): 213-219, 2020 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-31793637

RESUMEN

Mast cells (MCs) play a critical role in oral allergen-induced anaphylaxis. However, the contribution of basophils to the anaphylaxis remains unclear. The inhibitory immunoreceptor Allergin-1 is highly expressed on MCs and basophils and inhibits FcεRI-mediated signaling in MCs. Here, we show that Allergin-1-deficient (Milr1-/-) mice developed more severe hypothermia, a higher mortality rate and a greater incidence of diarrhea than did wild-type (WT) mice in an oral ovalbumin (OVA)-induced food allergy model. MC-deficient Mas-TRECK mice, which had been reconstituted with either WT or Milr1-/- bone marrow-derived cultured MCs, did not develop hypothermia in this food allergy model. On the other hand, depletion of basophils by injection of anti-CD200R3 antibody rescued Milr1-/- mice from lethal hypothermia but not from diarrhea. In vitro analyses demonstrated that Allergin-1 inhibits IgE-dependent activation of both human and mouse basophils. Thus, Allergin-1 on basophils selectively suppresses oral allergen-induced anaphylaxis.


Asunto(s)
Anafilaxia/inmunología , Basófilos/inmunología , Receptores Inmunológicos/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/deficiencia
4.
Nat Commun ; 10(1): 1365, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30911003

RESUMEN

Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial surface receptor genetically linked to the risk for Alzheimer's disease (AD). A proteolytic product, soluble TREM2 (sTREM2), is abundant in the cerebrospinal fluid and its levels positively correlate with neuronal injury markers. To gain insights into the pathological roles of sTREM2, we studied sTREM2 in the brain of 5xFAD mice, a model of AD, by direct stereotaxic injection of recombinant sTREM2 protein or by adeno-associated virus (AAV)-mediated expression. We found that sTREM2 reduces amyloid plaque load and rescues functional deficits of spatial memory and long-term potentiation. Importantly, sTREM2 enhances microglial proliferation, migration, clustering in the vicinity of amyloid plaques and the uptake and degradation of Aß. Depletion of microglia abolishes the neuroprotective effects of sTREM2. Our study demonstrates a protective role of sTREM2 against amyloid pathology and related toxicity and suggests that increasing sTREM2 can be explored for AD therapy.


Asunto(s)
Enfermedad de Alzheimer/terapia , Potenciación a Largo Plazo/efectos de los fármacos , Glicoproteínas de Membrana/genética , Microglía/efectos de los fármacos , Placa Amiloide/terapia , Receptores Inmunológicos/genética , Memoria Espacial/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inyecciones Intraventriculares , Potenciación a Largo Plazo/fisiología , Masculino , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/metabolismo , Ratones , Microglía/metabolismo , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fenotipo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Cultivo Primario de Células , Proteolisis , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Memoria Espacial/fisiología , Técnicas Estereotáxicas
6.
J Control Release ; 279: 326-335, 2018 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-29679665

RESUMEN

Exosomes are small membrane vesicles secreted by most cell types that play an important role in intercellular communication. Due to the characteristic of transferring their biomacromolecules, exosomes have potential as a new alternative for delivering protein therapeutics. Here, we investigate whether exosomes provide crucial advantages over other nanoparticles, in particular protein nanocage formulations, as a delivery system for membrane protein therapeutics. We characterized membrane-scaffold-based exosomes and protein-scaffold-based ferritin nanocages, both harboring SIRPα (signal regulatory protein α), an antagonist of CD47 on tumor cells. The efficacy of these two systems in delivering protein therapeutics was compared by testing their ability to enhance phagocytosis of tumor cells by bone-marrow-derived macrophages and subsequent inhibition of in vivo tumor growth. These analyses allowed us to comprehensively conclude that the therapeutic index of exosome-mediated CD47 blockade against tumor growth inhibition was higher than that of the same dose of ferritin-SIRPα. The results of this analysis reveal the importance of the unique characteristics of exosomes, in particular their membrane scaffold, in improving therapeutic protein delivery compared with protein-scaffold-based nanocages.


Asunto(s)
Antígenos de Diferenciación/administración & dosificación , Antígeno CD47/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos , Nanopartículas , Receptores Inmunológicos/administración & dosificación , Animales , Antígenos de Diferenciación/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Exosomas/química , Ferritinas/química , Células HT29 , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fagocitosis , Receptores Inmunológicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Am Heart Assoc ; 7(3)2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29378731

RESUMEN

BACKGROUND: Ischemic stroke is a complex disease with multiple etiologies and clinical manifestations. Paired immunoglobulin-like receptor B (PirB), which is originally thought to function exclusively in the immune system, is now also known to be expressed by neurons. A growing number of studies indicate that PirB can inhibit neurite outgrowth and restrict neuronal plasticity. The aim of the study is to investigate whether PirB can be an attractive theranostic target for ischemic stroke. METHODS AND RESULTS: First, we investigated the spatial-temporal expression of PirB in multiple ischemic stroke models, including transient middle cerebral artery occlusion, photothrombotic cerebral cortex ischemia, and the neuronal oxygen glucose deprivation model. Then, anti-PirB immunoliposome nanoprobe was developed by thin-film hydration method and investigated its specific targeting in vitro and in vivo. Finally, soluble PirB ectodomain (sPirB) protein delivered by polyethylene glycol-modified nanoliposome was used as a therapeutic reagent for ischemic stroke by blocking PirB binding to its endogenous ligands. These results showed that PirB was significantly upregulated after cerebral ischemic injury in ischemic stroke models. Anti-PirB immunoliposome nanoprobe was successfully developed and specifically bound to PirB in vitro. There was accumulation of anti-PirB immunoliposome nanoprobe in the ischemic hemisphere in vivo. Soluble PirB ectodomains remarkably improved ischemic stroke model recovery by liposomal delivery system. CONCLUSIONS: These data indicated that PirB was a significant element in the pathological process of cerebral ischemia. Therefore, PirB may act as a novel theranostic target for ischemic stroke.


Asunto(s)
Encéfalo/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/terapia , Imagen Molecular/métodos , Nanopartículas , Fragmentos de Péptidos/administración & dosificación , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/metabolismo , Espectroscopía Infrarroja Corta , Nanomedicina Teranóstica/métodos , Animales , Apoptosis , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Células Cultivadas , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ligandos , Liposomas , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores Inmunológicos/genética , Recuperación de la Función , Factores de Tiempo , Regulación hacia Arriba
8.
Neurobiol Dis ; 85: 164-173, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26550694

RESUMEN

BACKGROUND AND PURPOSE: Peripheral immune cell infiltration to the brain tissue at the perisurgical site can promote neuroinflammation after surgical brain injury (SBI). Slit2, an extracellular matrix protein, has been reported to reduce leukocyte migration. This study evaluated the effect of recombinant Slit2 and the role of its receptor roundabout1 (Robo1) and its downstream mediator Slit-Robo GTPase activating protein 1 (srGAP1)-Cdc42 on peripheral immune cell infiltration after SBI in a rat model. METHODS: One hundred and fifty-three adult male Sprague-Dawley rats (280-350 g) were used. Partial resection of right frontal lobe was performed to induce SBI. Slit2 siRNA was administered by intracerebroventricular injection 24h before SBI. Recombinant Slit2 was injected intraperitoneally 1h before SBI. Recombinant Robo1 used as a decoy receptor was co-administered with recombinant Slit2. srGAP1 siRNA was administered by intracerebroventricular injection 24h before SBI. Post-assessments included brain water content measurement, neurological tests, ELISA, Western blot, immunohistochemistry, and Cdc42 activity assay. RESULTS: Endogenous Slit2 was increased after SBI. Robo1 was expressed by peripheral immune cells. Endogenous Slit2 knockdown worsened brain edema after SBI. Recombinant Slit2 administration reduced brain edema, neurological deficits, and pro-inflammatory cytokines after SBI. Recombinant Slit2 reduced peripheral immune cell markers cluster of differentiation 45 (CD45) and myeloperoxidase (MPO), as well as Cdc42 activity in the perisurgical brain tissue which was reversed by recombinant Robo1 co-administration and srGAP1 siRNA. CONCLUSIONS: Recombinant Slit2 improved outcomes by reducing neuroinflammation after SBI, possibly by decreasing peripheral immune cell infiltration to the perisurgical site through Robo1-srGAP1 mediated inhibition of Cdc42 activity. These results suggest that Slit2 may be beneficial to reduce SBI-induced neuroinflammation.


Asunto(s)
Lesiones Encefálicas/inmunología , Lóbulo Frontal/inmunología , Lóbulo Frontal/lesiones , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Complicaciones Intraoperatorias/inmunología , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Animales , Edema Encefálico/etiología , Edema Encefálico/inmunología , Edema Encefálico/terapia , Lesiones Encefálicas/etiología , Lesiones Encefálicas/terapia , Modelos Animales de Enfermedad , Lóbulo Frontal/cirugía , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Infusiones Intraventriculares , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Complicaciones Intraoperatorias/terapia , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas Sprague-Dawley , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/genética , Proteínas Roundabout
9.
J Immunol ; 193(3): 1504-11, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973440

RESUMEN

We generated a new humanized mouse model to study HLA-restricted immune responses. For this purpose, we created unique murine hosts by enforcing the expression of human SIRPα by murine phagocytes in murine MHC-deficient HLA-transgenic alymphoid hosts, an approach that allowed the immune reconstitution of nonpermissive mice following injection of human hematopoietic stem cells. We showed that these mouse/human chimeras were able to generate HLA-restricted responses to immunization. These new humanized mice may offer attractive models to study immune responses to human diseases, such as HIV and EBV infections, as well as to assay new vaccine strategies.


Asunto(s)
Antígenos HLA/administración & dosificación , Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Quimera por Radiación/inmunología , Animales , Animales Recién Nacidos , Antígenos de Diferenciación/administración & dosificación , Antígenos de Diferenciación/sangre , Antígenos de Diferenciación/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Antígenos HLA/genética , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Quimera por Radiación/genética , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/sangre , Receptores Inmunológicos/genética
10.
PLoS One ; 9(3): e88259, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24642901

RESUMEN

Elucidating the sites and mechanisms of sRAGE action in the healthy state is vital to better understand the biological importance of the receptor for advanced glycation end products (RAGE). Previous studies in animal models of disease have demonstrated that exogenous sRAGE has an anti-inflammatory effect, which has been reasoned to arise from sequestration of pro-inflammatory ligands away from membrane-bound RAGE isoforms. We show here that sRAGE exhibits in vitro binding with high affinity and reversibly to extracellular matrix components collagen I, collagen IV, and laminin. Soluble RAGE administered intratracheally, intravenously, or intraperitoneally, does not distribute in a specific fashion to any healthy mouse tissue, suggesting against the existence of accessible sRAGE sinks and receptors in the healthy mouse. Intratracheal administration is the only effective means of delivering exogenous sRAGE to the lung, the organ in which RAGE is most highly expressed; clearance of sRAGE from lung does not differ appreciably from that of albumin.


Asunto(s)
Colágeno Tipo IV/metabolismo , Colágeno Tipo I/metabolismo , Laminina/metabolismo , Receptores Inmunológicos/metabolismo , Administración por Inhalación , Animales , Disponibilidad Biológica , Fibronectinas/metabolismo , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Cinética , Pulmón/química , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/aislamiento & purificación , Solubilidad
11.
J Innate Immun ; 6(3): 284-92, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24192271

RESUMEN

Activation of complement may cause severe tissue damage in antibody-mediated allograft rejection and other antibody-mediated clinical conditions; therefore, novel potent complement inhibitors are needed. Previously, we described binding of the inhibitory receptor LAIR-1 and its soluble family member LAIR-2 to collagen. Here, we investigated binding of LAIR-1 and LAIR-2 to the complement proteins C1q and MBL, which both have collagen-like domains, and evaluated the effect of this binding on complement function. We demonstrate specific binding of recombinant LAIR proteins to both C1q and MBL. Surface plasmon resonance experiments showed that LAIR-2-Fc protein bound C1q and MBL with the highest affinity compared to LAIR-2-HIS. We, therefore, hypothesized that LAIR-2-Fc is a potent complement inhibitor. Indeed, LAIR-2-Fc inhibited C4 fixation to IgG or mannan, reduced activation of C4 by aggregated IgG in plasma and inhibited iC3b deposition on cells. Finally, LAIR-2-Fc inhibited complement-mediated lysis of cells sensitized with anti-HLA antibodies in an ex vivo model for antibody-mediated transplant rejection. Thus, LAIR-2-Fc is an effective novel complement inhibitor for the treatment and prevention of antibody-mediated allograft rejection and antibody-mediated clinical conditions.


Asunto(s)
Rechazo de Injerto/prevención & control , Enfermedades del Complejo Inmune/terapia , Inmunoterapia , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Complemento C1q/metabolismo , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Rechazo de Injerto/inmunología , Antígenos HLA/inmunología , Humanos , Enfermedades del Complejo Inmune/inmunología , Regiones Constantes de Inmunoglobulina/genética , Isoanticuerpos/metabolismo , Células K562 , Unión Proteica/efectos de los fármacos , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética
12.
J Mol Med (Berl) ; 91(12): 1369-81, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24132651

RESUMEN

UNLABELLED: Signaling of the receptor for advanced glycation end products (RAGE) has been implicated in the development of injury-elicited vascular complications. Soluble RAGE (sRAGE) acts as a decoy of RAGE and has been used to treat pathological vascular conditions in animal models. However, previous studies used a high dose of sRAGE produced in insect Sf9 cells (sRAGE(Sf9))and multiple injections to achieve the therapeutic outcome. Here, we explore whether modulation of sRAGE N-glycoform impacts its bioactivity and augments its therapeutic efficacy. We first profiled carbohydrate components of sRAGE produced in Chinese hamster Ovary cells (sRAGE(CHO)) to show that a majority of its N-glycans belong to sialylated complex types that are not shared by sRAGE(Sf9). In cell-based NF-κB activation and vascular smooth muscle cell (VSMC) migration assays, sRAGE(CHO) exhibited a significantly higher bioactivity relative to sRAGE(Sf9) to inhibit RAGE alarmin ligand-induced NF-κB activation and VSMC migration. We next studied whether this N-glycoform-associated bioactivity of sRAGE(CHO) is translated to higher in vivo therapeutic efficacy in a rat carotid artery balloon injury model. Consistent with the observed higher bioactivity in cell assays, sRAGE(CHO) significantly reduced injury-induced neointimal growth and the expression of inflammatory markers in injured vasculature. Specifically, a single dose of 3 ng/g of sRAGE(CHO) reduced neointimal hyperplasia by over 70%, whereas the same dose of sRAGE(Sf9) showed no effect. The administered sRAGE(CHO) is rapidly and specifically recruited to the injured arterial locus, suggesting that early intervention of arterial injury with sRAGE(CHO) may offset an inflammatory circuit and reduce the ensuing tissue remodeling. Our findings showed that the N-glycoform of sRAGE is the key determinant underlying its bioactivity and thus is an important glycobioengineering target to develop a highly potent therapeutic sRAGE for future clinical applications. KEY MESSAGE: The specific N-glycoform modification is the key underlying sRAGE bioactivity Markedly reduced sRAGE dose to attenuate neointimal hyperplasia and inflammation Provide a molecular target for glycobioengineering of sRAGE as a therapeutic protein Blocking RAGE alarmin ligands during acute injury phase offsets neointimal growth.


Asunto(s)
Artritis/metabolismo , Artritis/patología , Neointima/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Artritis/tratamiento farmacológico , Biomarcadores/metabolismo , Células CHO , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Movimiento Celular/efectos de los fármacos , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Glicosilación , Humanos , Ligandos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , FN-kappa B/metabolismo , Neointima/tratamiento farmacológico , Ratas , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Células Sf9
13.
Mol Med ; 19: 183-94, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23821362

RESUMEN

Rheumatoid arthritis (RA) is a chronic inflammatory synovitis that leads to the destruction of bone and cartilage. The receptor for advanced glycation end products (RAGE) is a multiligand membrane-bound receptor for high-mobility group box-1 (HMGB1) associated with development of RA by inducing production of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1 and IL-6. We developed a bone-targeting therapeutic agent by tagging acidic oligopeptide to a nonmembrane-bound form of RAGE (endogenous secretory RAGE [esRAGE]) functioning as a decoy receptor. We assessed its tissue distribution and therapeutic effectiveness in a murine model of collagen-induced arthritis (CIA). Acidic oligopeptide-tagged esRAGE (D6-esRAGE) was localized to mineralized region in bone, resulting in the prolonged retention of more than 1 wk. Weekly administration of D6-esRAGE with a dose of 1 mg/kg to RA model mice significantly ameliorated inflammatory arthritis, synovial hyperplasia, cartilage destruction and bone destruction, while untagged esRAGE showed little effectiveness. Moreover, D6-esRAGE reduced plasma levels of proinflammatory cytokines including TNF-α, IL-1 and IL-6, while esRAGE reduced the levels of IL-1 and IL-6 to a lesser extent, suggesting that production of IL-1 and IL-6 reduced along the blockade of HMGB1 receptor downstream signals by D6-esRAGE could be attributed to remission of CIA. These findings indicate that D6-esRAGE enhances drug delivery to bone, leading to rescue of clinical and pathological lesions in murine CIA.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Huesos/metabolismo , Sistemas de Liberación de Medicamentos , Oligopéptidos/administración & dosificación , Receptores Inmunológicos/administración & dosificación , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Artritis Reumatoide , Ácido Aspártico/química , Línea Celular , Colágeno Tipo II/inmunología , Citocinas/sangre , Proteína HMGB1/farmacología , Masculino , Ratones , Ratones Endogámicos DBA , Receptor para Productos Finales de Glicación Avanzada
14.
Arterioscler Thromb Vasc Biol ; 33(3): 585-94, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288172

RESUMEN

OBJECTIVE: Little is known about the involvement of the soluble form of receptor for advanced glycation end products (sRAGE) in acute ischemic stroke (IS). Here, we aim to identify the role of plasma sRAGE and high mobility group box 1 (HMGB1) in imaging-confirmed IS patients, as well as mice subjected to focal ischemic stroke. METHODS AND RESULTS: IS patients were recruited and plasma samples were collected for the measurement of sRAGE and HMGB1 after stroke. The relation of sRAGE and HMGB1 with acute IS was also investigated in a C57BL/6J mouse model of focal ischemic stroke and primary cortical neurons subjected to oxygen and glucose deprivation. Plasma levels of sRAGE and HMGB1 were both significantly increased within 48 hours after IS, and the sRAGE level was an independent predictor of functional outcome at 3 months poststroke. Immunoprecipitation assays revealed that the binding of plasma HMGB1 to sRAGE increased progressively after IS both in patients and mice. Administration of recombinant sRAGE significantly reduced infiltrating immune cells and improved the outcome of injury in mice, protected cultured neurons against oxygen and glucose deprivation-induced cell death, and ameliorated the detrimental effect of recombinant HMGB1. CONCLUSIONS: Early poststroke plasma sRAGE may play a protective role in IS by capturing HMGB1. Hence, recombinant sRAGE is a potential therapeutic agent in acute IS.


Asunto(s)
Corteza Cerebral/metabolismo , Infarto de la Arteria Cerebral Media/sangre , Neuronas/metabolismo , Receptores Inmunológicos/sangre , Accidente Cerebrovascular/sangre , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Animales , Biomarcadores/sangre , Estudios de Casos y Controles , Muerte Celular , Hipoxia de la Célula , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Distribución de Chi-Cuadrado , Modelos Animales de Enfermedad , Femenino , Glucosa/deficiencia , Proteína HMGB1/sangre , Humanos , Inmunoprecipitación , Infarto de la Arteria Cerebral Media/diagnóstico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Modelos Logísticos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Oxígeno/metabolismo , Pronóstico , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Factores de Riesgo , Accidente Cerebrovascular/diagnóstico , Rehabilitación de Accidente Cerebrovascular , Taiwán , Factores de Tiempo , Regulación hacia Arriba
15.
Cell Tissue Res ; 351(1): 139-48, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23086479

RESUMEN

Receptor for advanced glycation end products (RAGE) is a pattern recognition receptor that binds a variety of pro-inflammatory ligands. Its soluble form, sRAGE, can compete for ligand binding and thereby have an anti-inflammatory effect. We have recently reported that sRAGE also exerts pro-inflammatory and chemotactic properties suggesting a dual role for sRAGE in immune modulation. Our present aim was to analyse the immunomodulatory properties of sRAGE in vivo with respect to acquired immunity. Naive mice were treated intra-peritoneally with sRAGE and cells from peritoneal lavage, spleens and bone marrow were examined. Mice treated with sRAGE displayed an increased leucocyte count in the peritoneal cavity, enlarged spleens and increased cellularity compared with vehicle-treated animals. Furthermore, sRAGE-treated mice had a significantly increased frequency and number of CD19(+) B cells in spleen and a reduced frequency of CD19(+) B cells in bone marrow compared with controls. Functionally, splenocytes from sRAGE-treated mice showed elevated IgG production and up to a four-fold increased IgM secretion compared with control animals and produced significantly higher levels of interleukin-10, interferon-γ and interleukin-6 in response to lipopolysaccharide stimulation. Our results suggest that sRAGE has immunomodulatory properties, since intra-peritoneal administration of sRAGE into healthy mice leads to rearrangements in cellular composition in the bone marrow and spleen. Moreover, the administration of sRAGE directs B cells into the spleen and towards differentiation. Our novel findings indicate that sRAGE exerts an effect on the cells of adaptive immunity.


Asunto(s)
Antígenos CD19/metabolismo , Complejo CD3/metabolismo , Tejido Linfoide/patología , Antígeno de Macrófago-1/metabolismo , Cavidad Peritoneal/patología , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/metabolismo , Animales , Formación de Anticuerpos/efectos de los fármacos , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/sangre , Femenino , Inyecciones Intraperitoneales , Leucocitos/citología , Lipopolisacáridos/farmacología , Tejido Linfoide/efectos de los fármacos , Ratones , Receptor para Productos Finales de Glicación Avanzada , Solubilidad , Bazo/efectos de los fármacos , Bazo/patología , Esplenomegalia/patología
16.
J Bone Miner Res ; 26(5): 1084-98, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21542009

RESUMEN

Alzheimer's disease (AD), one of the most dreaded neurodegenerative disorders, is characterized by cortical and cerebrovascular amyloid ß peptide (Aß) deposits, neurofibrillary tangles, chronic inflammation, and neuronal loss. Increased bone fracture rates and reduced bone density are commonly observed in patients with AD, suggesting one or more common denominators between both disorders. However, very few studies are available that have addressed this issue. Here, we present evidence for a function of amyloid precursor protein (APP) and Aß in regulating osteoclast (OC) differentiation in vitro and in vivo. Tg2576 mice, which express the Swedish mutation of APP (APPswe) under the control of a prion promoter, exhibit biphasic effects on OC activation, with an increase of OCs in younger mice (< 4 months old), but a decrease in older Tg2576 mice (> 4 months old). The increase of OCs in young Tg2576 mice appears to be mediated by Aß oligomers and receptor for advanced glycation end products (RAGE) expression in bone marrow macrophages (BMMs). However, the decrease of OC formation and activity in older Tg2576 mice may be due to the increase of soluble rage (sRAGE) in aged Tg2576 mice, an inhibitor of RANKL-induced osteoclastogenesis. These results suggest an unexpected function of APPswe/Aß, reveal a mechanism underlying altered bone remodeling in AD patients, and implicate APP/Aß and RAGE as common denominators for both AD and osteoporosis.


Asunto(s)
Envejecimiento/metabolismo , Péptidos beta-Amiloides/farmacología , Mutación/genética , Osteoblastos/citología , Osteoblastos/metabolismo , Receptores Inmunológicos/metabolismo , Envejecimiento/efectos de los fármacos , Péptidos beta-Amiloides/química , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Huesos/metabolismo , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Transgénicos , Modelos Biológicos , Tamaño de los Órganos , Especificidad de Órganos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Estructura Cuaternaria de Proteína , Ligando RANK/farmacología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Solubilidad/efectos de los fármacos
17.
J Immunol ; 186(7): 4051-8, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21346231

RESUMEN

Functional CD8 T cell effector and memory responses are generated and maintained during murine γ-herpesvirus 68 (γHV68) persistent infection despite continuous presentation of viral lytic Ags. However, the identity of the CD8 T cell subpopulations that mediate effective recall responses and that can participate in the generation of protective memory to a γ-herpesvirus infection remains unknown. During γHV68 persistence, ∼75% of γHV68-specific CD8 T cells coexpress the NK receptors killer cell lectin-like receptor G1 (KLRG1) and NKG2A. In this study, we take advantage of this unique phenotype to analyze the capacity of CD8 T cells expressing or not expressing KLRG1 and NKG2A to mediate effector and memory responses. Our results show that γHV68-specific KLRG1(+)NKG2A(+) CD8 T cells have an effector memory phenotype as well as characteristics of polyfunctional effector cells such us IFN-γ and TNF-α production, killing capacity, and are more efficient at protecting against a γHV68 challenge than their NKG2A(-)KLRG1(-) counterparts. Nevertheless, γHV68-specific NKG2A(+)KLRG1(+) CD8 T cells express IL-7 and IL-15 receptors, can survive long-term without cognate Ag, and subsequently mount a protective response during antigenic recall. These results highlight the plasticity of the immune system to generate protective effector and proliferative memory responses during virus persistence from a pool of KLRG1(+)NKG2A(+) effector memory CD8 T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/prevención & control , Memoria Inmunológica , Subfamília C de Receptores Similares a Lectina de Células NK/biosíntesis , Receptores Inmunológicos/biosíntesis , Rhadinovirus/inmunología , Infecciones Tumorales por Virus/prevención & control , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/trasplante , Linfocitos T CD8-positivos/virología , Proliferación Celular , Citotoxicidad Inmunológica , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Inmunofenotipificación , Lectinas Tipo C , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Subfamília C de Receptores Similares a Lectina de Células NK/administración & dosificación , Subfamília C de Receptores Similares a Lectina de Células NK/fisiología , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/fisiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/trasplante , Subgrupos de Linfocitos T/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/patología , Latencia del Virus/inmunología
18.
J Immunol ; 185(3): 1822-35, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20574008

RESUMEN

The receptor for advanced glycation end products (RAGE) is produced either as a transmembrane or soluble form (sRAGE). Substantial evidence supports a role for RAGE and its ligands in disease. sRAGE is reported to be a competitive, negative regulator of membrane RAGE activation, inhibiting ligand binding. However, some reports indicate that sRAGE is associated with inflammatory disease. We sought to define the biological function of sRAGE on inflammatory cell recruitment, survival, and differentiation in vivo and in vitro. To test the in vivo impact of sRAGE, the recombinant protein was intratracheally administered to mice, which demonstrated monocyte- and neutrophil-mediated lung inflammation. We also observed that sRAGE induced human monocyte and neutrophil migration in vitro. Human monocytes treated with sRAGE produced proinflammatory cytokines and chemokines. Our data demonstrated that sRAGE directly bound human monocytes and monocyte-derived macrophages. Binding of sRAGE to monocytes promoted their survival and differentiation to macrophages. Furthermore, sRAGE binding to cells increased during maturation, which was similar in freshly isolated mouse monocytes compared with mature tissue macrophages. Because sRAGE activated cell survival and differentiation, we examined intracellular pathways that were activated by sRAGE. In primary human monocytes and macrophages, sRAGE treatment activated Akt, Erk, and NF-kappaB, and their activation appeared to be critical for cell survival and differentiation. Our data suggest a novel role for sRAGE in monocyte- and neutrophil-mediated inflammation and mononuclear phagocyte survival and differentiation.


Asunto(s)
Diferenciación Celular/inmunología , Productos Finales de Glicación Avanzada/metabolismo , Monocitos/citología , Monocitos/inmunología , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/fisiología , Animales , Caspasa 3/metabolismo , Inhibidores de Caspasas , Adhesión Celular/inmunología , Línea Celular Tumoral , Supervivencia Celular/inmunología , Regulación hacia Abajo/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Monocitos/patología , FN-kappa B/biosíntesis , FN-kappa B/metabolismo , Infiltración Neutrófila/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Solubilidad , Regulación hacia Arriba/inmunología
19.
Curr Drug Targets ; 11(7): 875-81, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20412043

RESUMEN

Accelerated atherosclerosis is the leading cause of coronary heart disease and stroke, which could account for high mortality rates in patients with diabetes. Although several hyperglycemia-elicited metabolic and hemodynamic derangements have been implicated in the pathogenesis of cardiovascular disease (CVD) in diabetes, the process of formation and accumulation of advanced glycation end products (AGEs) and their mode of action are most compatible with the phenomenon 'metabolic memory' and 'legacy effect', that is, vascular stresses during the diabetic exposure have persisted after glucose normalization. Further, there is a growing body of evidence that a receptor for AGEs (RAGE) is involved in signal transduction of AGEs in a variety of cells. In this paper, we review the role of the AGE-RAGE system in accelerated atherosclerosis, especially focusing on smooth muscle cell pathophysiology, and also discuss the possibility that the AGE/RAGE axis could be a potential therapeutic target for prevention of CVD in patients with diabetes.


Asunto(s)
Aterosclerosis/metabolismo , Enfermedades Cardiovasculares/prevención & control , Angiopatías Diabéticas/prevención & control , Sistemas de Liberación de Medicamentos/métodos , Productos Finales de Glicación Avanzada/metabolismo , Hipoglucemiantes/farmacología , Miocitos del Músculo Liso/metabolismo , Aterosclerosis/complicaciones , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/metabolismo , Angiopatías Diabéticas/complicaciones , Angiopatías Diabéticas/metabolismo , Humanos , Hipoglucemiantes/administración & dosificación , Modelos Cardiovasculares , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/efectos de los fármacos , Receptores Inmunológicos/metabolismo
20.
Crit Care Med ; 38(6): 1414-22, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20386310

RESUMEN

OBJECTIVE: The receptor for advanced glycation end products mediates a variety of inflammatory responses. Soluble receptor for advanced glycation end products has been suggested to function as a decoy abrogating cellular activation. High-mobility group box 1 is a high-affinity binding ligand for the receptor for advanced glycation end products with cytokine activities and plays a role in sepsis. DESIGN: Controlled, in vivo laboratory study. SETTING: Research laboratory of a health sciences university. SUBJECTS: C57BL/6 mice. INTERVENTIONS: Peritonitis was induced by intraperitoneal injection of Escherichia coli. Mice received soluble receptor for advanced glycation end products or anti-high-mobility group box 1 immunoglobulin G, or the appropriate control treatment. MEASUREMENTS AND MAIN RESULTS: Soluble receptor for advanced glycation end products-treated mice demonstrated an enhanced bacterial dissemination to liver and lungs, accompanied by increased hepatocellular injury and exaggerated systemic cytokine release, 20 hrs after intraperitoneal administration of Escherichia coli. Soluble receptor for advanced glycation end products administration in healthy, uninfected mice did not induce an immune response. Remarkably, lung inflammation was unaffected. Furthermore, high-mobility group box 1 release was enhanced during peritonitis and anti-high-mobility group box 1 treatment was associated with higher bacterial loads in liver and lungs. CONCLUSIONS: These data are the first to suggest that receptor for advanced glycation end products ligands, including high-mobility group box 1, limit bacterial dissemination during Gram-negative sepsis.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Proteína HMGB1/fisiología , Inmunidad Innata/fisiología , Peritonitis/inmunología , Receptores Inmunológicos/inmunología , Sepsis/inmunología , Animales , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/patología , Femenino , Proteína HMGB1/administración & dosificación , Infusiones Parenterales , Ligandos , Hígado/metabolismo , Hígado/microbiología , Hígado/patología , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Peritonitis/metabolismo , Peritonitis/patología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Sepsis/metabolismo , Sepsis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...