Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Environ Toxicol ; 38(2): 253-265, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36350155

RESUMEN

BACKGROUND: Allergic rhinitis (AR) is an immunoglobulin E (IgE)-mediated immune inflammatory response that mainly affects the nasal mucosa. Currently, there is evidence that apigenin, as a flavonoid, has anti-allergic potential. MATERIAL/METHODS: In vitro, compound 48/80 and lipopolysaccharide (LPS) were used to induce mast cell activation and inflammation in HMC-1 cells. In vivo, ovalbumin (OVA) induced and stimulated AR in BALB/c mice. ELISA was used to detect the contents of ß-hexosaminidase, histamine, eosinophil cationic protein (ECP), OVA-specific IgE, IgG1, and IgG2a, inflammatory factors in cells and mouse serum. Cell viability and apoptosis were measured with MTT and flow cytometry. Toll like receptor 4 (TLR4)/myeloid differentiation factor88 (MyD88)/Nuclear transcription factor-κB (NF-κB) pathway-related proteins in cells and mouse nasal mucosa tissues were analyzed with Western blotting. The levels of Th1 (IFN-γ) and Th2 (IL-4, IL-5, and IL-13) cytokines and Th1 (T-bet) and Th2 (GATA-3) specific transcription factors were also assessed. The ratio of Th1 (CD4+ IFN-γ+ ) / Th2 (CD4+ IL-4+ ) cells in mouse peripheral blood mononuclear cells was evaluated by flow cytometry. RESULTS: Apigenin significantly inhibited compound 48/80-induced secretion of ß-hexosaminidase and histamine. Apigenin blocked LPS-induced decrease in cell viability and increase in cell apoptosis and inflammatory cytokine secretion by suppressing the activity of the TLR4/MyD88/NF-κB pathway. Apigenin treatment reduced the levels of OVA-specific IgE, IgG1 and IgG2a as well as ß-hexosaminidase, histamine and ECP levels in mouse serum. Moreover, administration with apigenin decreased Th2 cytokine and transcription factor levels and increased Th1 cytokine and transcription factor levels, and promoted the ratio of Th1/Th2 cells in AR mice. Additionally, apigenin significantly alleviated nasal symptoms and nasal eosinophil infiltration in AR mice. CONCLUSIONS: Apigenin alleviates the inflammatory response of allergic rhinitis by inhibiting the activity of the TLR4/MyD88/NF-κB signaling pathway.


Asunto(s)
Apigenina , Factor 88 de Diferenciación Mieloide , FN-kappa B , Rinitis Alérgica , Receptor Toll-Like 4 , Animales , Ratones , Apigenina/farmacología , Apigenina/uso terapéutico , beta-N-Acetilhexosaminidasas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Histamina/toxicidad , Inmunoglobulina E , Inmunoglobulina G/toxicidad , Inmunoglobulina G/metabolismo , Interleucina-4 , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Ratones Endogámicos BALB C , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Ovalbúmina/farmacología , Rinitis Alérgica/inducido químicamente , Rinitis Alérgica/tratamiento farmacológico , Transducción de Señal , Células Th2 , Receptor Toll-Like 4/metabolismo
2.
Emerg Microbes Infect ; 10(1): 2194-2198, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34736354

RESUMEN

Inactivated coronaviruses, including severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) and Middle East respiratory syndrome coronavirus (MERS-CoV), as potential vaccines have been reported to result in enhanced respiratory diseases (ERDs) in murine and nonhuman primate (NHP) pneumonia models after virus challenge, which poses great safety concerns of antibody-dependent enhancement (ADE) for the rapid wide application of inactivated SARS-CoV-2 vaccines in humans, especially when the neutralizing antibody levels induced by vaccination or initial infection quickly wane to nonneutralizing or subneutralizing levels over the time. With passive transfer of diluted postvaccination polyclonal antibodies to mimic the waning antibody responses after vaccination, we found that in the absence of cellular immunity, passive infusion of subneutralizing or nonneutralizing anti-SARS-CoV-2 antibodies could still provide some level of protection against infection upon challenge, and no low-level antibody-enhanced infection was observed. The anti-SARS-CoV-2 IgG-infused group and control group showed similar, mild to moderate pulmonary immunopathology during the acute phase of virus infection, and no evidence of vaccine-related pulmonary immunopathology enhancement was found. Typical immunopathology included elevated MCP-1, IL-8 and IL-33 in bronchoalveolar lavage fluid; alveolar epithelial hyperplasia; and exfoliated cells and mucus in bronchioles. Our results corresponded with the recent observations that no pulmonary immunology was detected in preclinical studies of inactivated SARS-CoV-2 vaccines in either murine or NHP pneumonia models or in large clinical trials and further supported the safety of inactivated SARS-CoV-2 vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Inmunogenicidad Vacunal , SARS-CoV-2/inmunología , Células Epiteliales Alveolares/patología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/toxicidad , Bronquiolos/química , Bronquiolos/patología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , COVID-19/patología , COVID-19/virología , Citocinas/análisis , Humanos , Hiperplasia , Inmunoglobulina G/inmunología , Inmunoglobulina G/toxicidad , Pulmón/patología , Macaca mulatta , Masculino , Ratones , Moco , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Vacunas de Productos Inactivados/inmunología
3.
Neurotox Res ; 39(4): 1300-1309, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33999356

RESUMEN

Neuromyelitis optica (NMO) and myelin oligodendrocyte glycoprotein (MOG) antibody-related disease (MOG disease) are inflammatory demyelinating diseases of the central nervous system (CNS). The disruption of the blood-brain barrier (BBB) is considered a key step in the pathogenesis of NMO and MOG disease. Although a previous report indicated that circulating immunoglobulin G (IgG) from NMO patients disrupts the BBB, the effect of IgG from patients with MOG disease has not been elucidated. In addition, it has been reported that some disease-modifying drugs for multiple sclerosis are harmful to NMO by an unknown mechanism. This study aimed to examine the effects of IgG from patients with NMO or MOG disease on BBB integrity. We also examined the effects of disease-modifying drugs (fingolimod [FTY720] and dimethyl fumarate [DMF]) on IgG-treated brain capillary endothelial cells. We used in vitro BBB models constructed with rat brain capillary endothelial cells (RBECs) to examine the effects on BBB function. The integrity of the RBECs was assessed by measuring transendothelial resistance (TEER) and cell viability. NMO or MOG-IgG treatment decreased TEER and cell viability in the endothelial monolayer model. Although FTY720 and DMF did not affect barrier function or cell viability under normal conditions, disease IgG-induced barrier dysfunctions were worsened by the presence of FTY720. These data indicate that circulating IgG in patients with NMO or MOG disease worsens BBB function. Furthermore, in patients with NMO or MOG disease treated with FTY720, changes in the integrity of the BBB were found to exacerbate the disease.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Clorhidrato de Fingolimod/toxicidad , Inmunoglobulina G/toxicidad , Glicoproteína Mielina-Oligodendrócito/toxicidad , Neuromielitis Óptica , Moduladores de los Receptores de fosfatos y esfingosina 1/toxicidad , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/metabolismo , Ratas , Ratas Wistar
4.
J Neuroinflammation ; 17(1): 190, 2020 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546235

RESUMEN

BACKGROUND: Opsoclonus-myoclonus syndrome (OMS) is a rare neurological disease. Some children with OMS also have neuroblastoma (NB). We and others have previously documented that serum IgG from children with OMS and NB induces neuronal cytolysis and activates several signaling pathways. However, the mechanisms underlying OMS remain unclear. Here, we investigated whether nitric oxide (NO) from activated microglias and its cascade contribute to neuronal cytolysis in pediatric OMS. METHODS: The activation of cultured cerebral cortical and cerebellar microglias incubated with sera or IgG isolated from sera of children with OMS and NB was measured by the expression of the activation marker, cytokines, and NO. Neuronal cytolysis was determined after exposing to IgG-treated microglia-conditioned media. Using inhibitors and activators, the effects of NO synthesis and its intracellular cascade, namely soluble guanylyl cyclase (sGC) and protein kinase G (PKG), on neuronal cytolysis were evaluated. RESULTS: Incubation with sera or IgG from children with OMS and NB increased the activation of cerebral cortical and cerebellar microglias, but not the activation of astrocytes or the cytolysis of glial cells. Moreover, the cytolysis of neurons was elevated by conditioned media from microglias incubated with IgG from children with OMS and NB. Furthermore, the expression of NO, sGC, and PKG was increased. Neuronal cytolysis was relieved by the inhibitors of NO signaling, while neuronal cytolysis was exacerbated by the activators of NO signaling but not proinflammatory cytokines. The cytolysis of neurons was suppressed by pretreatment with the microglial inhibitor minocycline, a clinically tested drug. Finally, increased microglial activation did not depend on the Fab fragment of serum IgG. CONCLUSIONS: Serum IgG from children with OMS and NB potentiates microglial activation, which induces neuronal cytolysis through the NO/sGC/PKG pathway, suggesting an applicability of microglial inhibitor as a therapeutic candidate.


Asunto(s)
Inmunoglobulina G/toxicidad , Microglía/efectos de los fármacos , Neuroblastoma/complicaciones , Neuronas/patología , Síndrome de Opsoclonía-Mioclonía/inmunología , Niño , Preescolar , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Femenino , Guanilato Ciclasa/metabolismo , Humanos , Inmunoglobulina G/inmunología , Masculino , Microglía/inmunología , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuronas/efectos de los fármacos , Óxido Nítrico/metabolismo , Síndrome de Opsoclonía-Mioclonía/etiología , Síndrome de Opsoclonía-Mioclonía/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
5.
J Neuroinflammation ; 17(1): 31, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973738

RESUMEN

OBJECTIVE: Autoantibodies against ribosomal P proteins (anti-P antibodies) are strongly associated with the neuropsychiatric manifestations of systemic lupus erythematosus (NPSLE). The present study was designed to assess whether anti-P antibodies can induce abnormal brain electrical activities in mice and investigate the potential cytopathological mechanism. METHODS: Affinity-purified human anti-ribosomal P antibodies were injected intravenously into mice after blood-brain barrier (BBB) disruption. The auditory steady-state response (ASSR) was evaluated based on electroencephalography (EEG) signals in response to 40-Hz click-train stimuli, which were recorded from electrodes implanted in the skull of mice. Immunofluorescence staining was used to examine the morphology and density of neurons and glia in the hippocampus and cortex. The presence of apoptosis in the brain tissues was studied using the TUNEL assay. A PLX3397 diet was used to selectively eliminate microglia from the brains of mice. RESULTS: Circulating anti-P antibodies caused an enhancement of the ASSR and the activation of microglia through the disrupted BBB, while no obvious neural apoptosis was observed. In contrast, when microglia were depleted, anti-P antibodies induced a serious reduction in the ASSR and neural apoptosis. CONCLUSION: Our study indicates that anti-P antibodies can directly induce the dysfunction of auditory-evoked potentials in the brain and that microglia are involved in the protection of neural activity after the invasion of anti-P antibodies, which could have important implications for NPSLE.


Asunto(s)
Autoanticuerpos/toxicidad , Potenciales Evocados Auditivos/efectos de los fármacos , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Microglía/inmunología , Proteínas Ribosómicas/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL
6.
J Pharm Sci ; 109(1): 761-768, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31376374

RESUMEN

Closed system transfer devices (CSTD) are a supplemental engineering control designed to reduce occupational exposure of hazardous drugs and are currently implemented in accordance with evolving regulations. Owing to the novelty and complexity of these devices and their importance in clinical in-use testing, here we evaluated FDA-approved CSTD, assessing product quality through stability indicating assays to determine any drug product incompatibilities. Six devices were used in a simulated compounding and administration of a late-phase IgG1 antibody-drug conjugate (ADC) and the resulting samples were analyzed for visible and subvisible particle counts by light obscuration and micro-flow imaging, physical stability by size exclusion chromatography, and biological activities by relative potency. Potential challenges included improper fit of CSTD components, loss of product to void volume, and material incompatibility. Results showed compatibility of the ADC with the 6 CSTD evaluated. One CSTD introduced subvisible particles into the ADC during compounding that were identified through morphological assessment as silicone oil. This study highlights the importance of clinical in use testing with new devices and proposes strategies to mitigate the risk of drug product incompatibility with CSTD.


Asunto(s)
Composición de Medicamentos/instrumentación , Inmunoconjugados/química , Inmunoglobulina G/química , Exposición Profesional/prevención & control , Equipos de Seguridad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Estabilidad de Medicamentos , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/toxicidad , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/toxicidad , Ensayo de Materiales , Exposición Profesional/efectos adversos , Agregado de Proteínas , Estabilidad Proteica
7.
Eur J Pharm Biopharm ; 146: 73-83, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31811896

RESUMEN

Polysorbates (PSs) are common protein stabilizers used in biotherapeutic formulations. However, PSs are heterogeneous and unstable in liquid protein formulations [1,2]. The purpose of this work is to explore possible alternatives for polysorbate replacements that demonstrate superior protein protection, superior self-stability, low toxicity, and wide applicability. For this purpose, 8 non-ionic surfactants that have not yet been used as excipients in marketed biotherapeutic products were investigated with PS20/80 as the benchmark. Compared with PS20/80, Brij-58 showed better protein protection ability in the mAb1 formulation under forced degradation conditions when examined by visual inspection, SEC, and dynamic lighting scanning. Additionally, Brij-58 has a better inherent stability than PS20/80 in the protein formulation when detected by UPLC-CAD. Moreover, Brij-58 is an inert excipient that does not affect protein bioactivity and conformation. In addition, the LD50 and hemolysis concentration of Brij-58 were determined, which is relatively safe when used as a parenteral injection. Furthermore, Brij-58 was also an effective protein stabilizer for the other two antibody products (IgG4 subtype and bispecific antibody) in the shaking study. In summary, Brij-58 stands out as a promising PS replacement in biotherapeutic formulations with a safe, stable and effective protein-protection profile among candidate surfactants.


Asunto(s)
Productos Biológicos/química , Cetomacrogol/química , Composición de Medicamentos/métodos , Excipientes/química , Tensoactivos/química , Administración Intravenosa , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/toxicidad , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/toxicidad , Productos Biológicos/administración & dosificación , Productos Biológicos/toxicidad , Cetomacrogol/toxicidad , Química Farmacéutica , Estabilidad de Medicamentos , Excipientes/toxicidad , Femenino , Células HEK293 , Hemólisis/efectos de los fármacos , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/química , Inmunoglobulina G/toxicidad , Dosificación Letal Mediana , Masculino , Ratones , Polisorbatos/química , Polisorbatos/toxicidad , Estabilidad Proteica , Conejos , Tensoactivos/toxicidad , Pruebas de Toxicidad Aguda
8.
Neurosci Bull ; 35(5): 853-866, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31041694

RESUMEN

Immunoglobulin G against myelin oligodendrocyte glycoprotein (MOG-IgG) is detectable in neuromyelitis optica spectrum disorder (NMOSD) without aquaporin-4 IgG (AQP4-IgG), but its pathogenicity remains unclear. In this study, we explored the pathogenic mechanisms of MOG-IgG in vitro and in vivo and compared them with those of AQP4-IgG. MOG-IgG-positive serum induced complement activation and cell death in human embryonic kidney (HEK)-293T cells transfected with human MOG. In C57BL/6 mice and Sprague-Dawley rats, MOG-IgG only caused lesions in the presence of complement. Interestingly, AQP4-IgG induced astroglial damage, while MOG-IgG mainly caused myelin loss. MOG-IgG also induced astrocyte damage in mouse brains in the presence of complement. Importantly, we also observed ultrastructural changes induced by MOG-IgG and AQP4-IgG. These findings suggest that MOG-IgG directly mediates cell death by activating complement in vitro and producing NMOSD-like lesions in vivo. AQP4-IgG directly targets astrocytes, while MOG-IgG mainly damages oligodendrocytes.


Asunto(s)
Acuaporina 4/toxicidad , Astrocitos/patología , Proteínas del Sistema Complemento , Inmunoglobulina G/toxicidad , Glicoproteína Mielina-Oligodendrócito/toxicidad , Oligodendroglía/patología , Adolescente , Adulto , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Niño , Proteínas del Sistema Complemento/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Ratas , Ratas Sprague-Dawley , Adulto Joven
9.
Pharm Res ; 36(3): 47, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-30721414

RESUMEN

PURPOSE: The Göttingen minipig is a relevant non-rodent species for regulatory toxicological studies. Yet, its use with therapeutic antibodies has been limited by the unknown binding properties of human immunoglobulins (huIgG) to porcine Fc gamma receptors (poFcγR) influencing safety and efficacy readouts. Therefore, knowing IgG-FcγR interactions in the animal model is a prerequisite for the use of minipigs in preclinical safety and efficacy studies with therapeutic antibodies. METHODS: Here, we describe the cloning and expression of poFcγRs and their interactions with free and complexed human therapeutic IgG1 by surface plasmon resonance and flow cytometry. RESULTS: We show here that poFcγRIa, poFcγRIIa, and poFcγRIIb bind huIgG1 antibodies with comparable affinities as corresponding huFcγRs. Importantly, poFcγRs bind huIgG immune complexes with high avidity, thus probably allowing human-like effector functions. However, poFcγRIIIa binds poIgG1a but not to huIgG1. CONCLUSIONS: The lack of binding of poFcγRIIIa to huIgG1 might cause underestimation of FcγRIIIa-mediated efficacy or toxicity as mediated by porcine natural killer cells. Therefore, the suitability of minipigs in preclinical studies with human therapeutic antibodies has to be assessed case by case. Our results facilitate the use of Göttingen minipigs for assessment of human therapeutic antibodies in preclinical studies.


Asunto(s)
Inmunoglobulina G/metabolismo , Receptores de IgG/metabolismo , Animales , Afinidad de Anticuerpos , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/toxicidad , Células Asesinas Naturales/metabolismo , Unión Proteica , Porcinos , Porcinos Enanos
10.
Glia ; 66(12): 2575-2588, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30240044

RESUMEN

Multiple sclerosis (MS) and neuromyelitis optica (NMO) are inflammatory demyelinating disorders of the central nervous system with evidence of antibody-mediated pathology. Using ex vivo organotypic mouse cerebellar slice cultures, we have demonstrated that recombinant antibodies (rAbs) cloned from cerebrospinal fluid plasmablasts of MS and NMO patients target myelin- and astrocyte-specific antigens to induce disease-specific oligodendrocyte loss and myelin degradation. In this study, we examined glial cell responses and myelin integrity during recovery from disease-specific antibody-mediated injury. Following exposure to MS rAb and human complement (HC) in cerebellar explants, myelinating oligodendrocytes repopulated the demyelinated tissue and formed new myelin sheaths along axons. Remyelination was accompanied by pronounced microglial activation. In contrast, following treatment with NMO rAb and HC, there was rapid regeneration of astrocytes and pre-myelinating oligodendrocytes but little formation of myelin sheaths on preserved axons. Deficient remyelination was associated with progressive axonal loss and the return of microglia to a resting state. Our results indicate that antibody-mediated demyelination in MS and NMO show distinct capacities for recovery associated with differential injury to adjacent axons and variable activation of microglia. Remyelination was rapid in MS rAb plus HC-induced demyelination. By contrast, oligodendrocyte maturation and remyelination failed following NMO rAb-mediated injury despite the rapid restoration of astrocytes and preservation of axons in early lesions.


Asunto(s)
Cerebelo , Inmunoglobulina G/toxicidad , Esclerosis Múltiple/inmunología , Neuroglía/efectos de los fármacos , Neuromielitis Óptica/inmunología , Remielinización/efectos de los fármacos , Animales , Animales Recién Nacidos , Acuaporina 4/metabolismo , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gutatión-S-Transferasa pi/metabolismo , Humanos , Ratones , Ratones Transgénicos , Esclerosis Múltiple/sangre , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/metabolismo , Neuroglía/fisiología , Neuromielitis Óptica/sangre , Técnicas de Cultivo de Órganos , Remielinización/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Factores de Transcripción SOXB1/metabolismo
11.
PLoS One ; 13(4): e0191926, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29617360

RESUMEN

CTLA-4 and CD28 exemplify a co-inhibitory and co-stimulatory signaling axis that dynamically sculpts the interaction of antigen-specific T cells with antigen-presenting cells. Anti-CTLA-4 antibodies enhance tumor-specific immunity through a variety of mechanisms including: blockade of CD80 or CD86 binding to CTLA-4, repressing regulatory T cell function and selective elimination of intratumoral regulatory T cells via an Fcγ receptor-dependent mechanism. AGEN1884 is a novel IgG1 antibody targeting CTLA-4. It potently enhanced antigen-specific T cell responsiveness that could be potentiated in combination with other immunomodulatory antibodies. AGEN1884 was well-tolerated in non-human primates and enhanced vaccine-mediated antigen-specific immunity. AGEN1884 combined effectively with PD-1 blockade to elicit a T cell proliferative response in the periphery. Interestingly, an IgG2 variant of AGEN1884 revealed distinct functional differences that may have implications for optimal dosing regimens in patients. Taken together, the pharmacological properties of AGEN1884 support its clinical investigation as a single therapeutic and combination agent.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antineoplásicos Inmunológicos/farmacología , Antígeno CTLA-4/inmunología , Inmunoglobulina G/farmacología , Neoplasias/terapia , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacocinética , Adyuvantes Inmunológicos/toxicidad , Secuencia de Aminoácidos , Animales , Formación de Anticuerpos/efectos de los fármacos , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/toxicidad , Células CHO , Antígeno CTLA-4/antagonistas & inhibidores , Vacunas contra el Cáncer/farmacología , Células Cultivadas , Cricetulus , Mapeo Epitopo , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunoglobulina G/química , Inmunoglobulina G/toxicidad , Activación de Linfocitos/efectos de los fármacos , Macaca fascicularis , Modelos Moleculares , Neoplasias/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
12.
Mol Immunol ; 85: 89-99, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28214650

RESUMEN

Increasing evidences indicate that 2-Methoxyestradiol (2ME2) plays an essential role in protecting against inflammatory responses. However, its effect on IgG immune complex (IC)-induced acute lung injury (ALI) remains enigmatic. In the study, by using i.p. administration of 2ME2, we evaluated its influence on IgG IC-induced pulmonary injury in mice. We found that during IgG IC-induced ALI, mice treated by 2ME2 displayed a substantial decrease in vascular permeability and neutrophil influx (represented by myeloperoxidase activity) when compared with their counterparts receiving vehicle treatment. Furthermore, 2ME2 treatment significantly decreased pro-inflammatory mediator production and inflammatory cell, especially neutrophil accumulation in bronchoalveolar lavage fluids (BALFs) upon IgG IC stimulation. In vitro, IgG IC-triggered inflammatory mediator production was markedly down-regulated by 2ME2 in macrophages. Moreover, we verified that the activation of the transcription factors, NF-κB and CCAAT/enhancer-binding protein (C/EBP) ß, were inhibited by 2ME2 in IgG IC-challenged macrophages. We demonstrated that alleviation of NF-κB-dependent transcription might be associated with reduced phosphorylation of NF-κB p65, and reduction of C/EBP activation was directly linked to its expression. In addition, we discovered that IgG IC-stimulated phosphorylation of both p38 MAPK and ERK1/2 was alleviated by 2ME2. These data indicated a novel strategy for blockade of IgG IC-induced inflammatory activities.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/efectos de los fármacos , Estradiol/análogos & derivados , Macrófagos Alveolares/efectos de los fármacos , FN-kappa B/efectos de los fármacos , 2-Metoxiestradiol , Lesión Pulmonar Aguda/inmunología , Animales , Complejo Antígeno-Anticuerpo/toxicidad , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/inmunología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Modelos Animales de Enfermedad , Estradiol/farmacología , Inmunoglobulina G/toxicidad , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
J Neuroinflammation ; 13(1): 275, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27765056

RESUMEN

BACKGROUND: Neuromyelitis optica (NMO), an autoimmune inflammatory disease of the central nervous system, is often associated with retinal abnormalities including thinning of the retinal nerve fiber layer and microcystic changes. Here, we demonstrate that passive transfer of an anti-aquaporin-4 autoantibody (AQP4-IgG) produces primary retinal pathology. METHODS: AQP4-IgG was delivered to adult rat retinas by intravitreal injection. Rat retinas and retinal explant cultures were assessed by immunofluorescence. RESULTS: Immunofluorescence showed AQP4-IgG deposition on retinal Müller cells, with greatly reduced AQP4 expression and increased glial fibrillary acidic protein by 5 days. There was mild retinal inflammation with microglial activation but little leukocyte infiltration and loss of retinal ganglion cells by 30 days with thinning of the ganglion cell complex. Interestingly, the loss of AQP4 was complement independent as seen in cobra venom factor-treated rats and in normal rats administered a mutated AQP4-IgG lacking complement effector function. Exposure of ex vivo retinal cultures to AQP4-IgG produced a marked reduction in AQP4 expression by 24 h, which was largely prevented by inhibitors of endocytosis or lysosomal acidification. CONCLUSIONS: Passive transfer of AQP4-IgG results in primary, complement-independent retinal pathology, which might contribute to retinal abnormalities seen in NMO patients.


Asunto(s)
Acuaporina 4/inmunología , Proteínas del Sistema Complemento/metabolismo , Inmunoglobulina G/toxicidad , Neuromielitis Óptica , Retina/metabolismo , Retina/patología , Animales , Acuaporina 4/metabolismo , Proteínas de Unión al Calcio/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inyecciones Intravítreas , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Masculino , Proteínas de Microfilamentos/metabolismo , Neuromielitis Óptica/etiología , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/patología , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología
14.
J Thromb Haemost ; 13(11): 2053-62, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26340698

RESUMEN

BACKGROUND: Acquired thrombotic thrombocytopenic purpura (TTP) is caused by an autoantibody-mediated deficiency of the von Willebrand factor-cleaving protease ADAMTS-13. Acute episodes of the disease are treated with a combination of immunosuppression and repeated cycles of plasma exchange to remove anti-ADAMTS-13 autoantibodies and, at the same time, replenish functional ADAMTS-13. Although this is often effective, the mortality rate has remained between 10% and 20%, highlighting the need for safer treatment options. OBJECTIVES: We previously showed that, in vitro, human recombinant ADAMTS-13 (rADAMTS-13) is able to override neutralizing antibodies and restore ADAMTS-13 activity in plasma from patients with acquired TTP. In the present study, we assessed the in vivo feasibility of this strategy by using a rat model. METHODS: Wild-type rats were adjusted to an ADAMTS-13 inhibitor (inhibitor) titer of ~ 10 BU mL(-1) with goat anti-ADAMTS-13 IgG, and treated with increasing doses of rADAMTS-13. Blood samples were drawn and analyzed for ADAMTS-13-specific parameters, including FRETS-VWF73 activity, inhibitor, and ADAMTS-13-specific immune complexes (ICs). The pharmacokinetics of ADAMTS-13 activity and inhibitors were evaluated. RESULTS: Administration of inhibitor titer-adjusted doses of rADAMTS-13 to inhibitor-treated rats predictably restored activity. Inhibitors were readily neutralized through formation of ADAMTS-13-specific ICs, which were cleared at a higher rate than the free inhibitor. Surplus protease was enzymatically active in plasma, and showed similar pharmacokinetics to ADAMTS-13 in not inhibitor-treated rats. CONCLUSIONS: Defined doses of rADAMTS-13 neutralized circulating anti-ADAMTS-13 antibodies and enabled reconstitution of ADAMTS-13 activity in plasma in our model, indicating that the protease may be a promising candidate for further exploration in treating acute episodes of acquired TTP.


Asunto(s)
Proteínas ADAM/uso terapéutico , Anticuerpos Neutralizantes/sangre , Autoanticuerpos/sangre , Púrpura Trombocitopénica Trombótica/inmunología , Proteínas ADAM/sangre , Proteínas ADAM/deficiencia , Proteínas ADAM/inmunología , Proteína ADAMTS13 , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/toxicidad , Complejo Antígeno-Anticuerpo/sangre , Autoanticuerpos/inmunología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Cabras/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina G/toxicidad , Masculino , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Factor de von Willebrand/metabolismo
15.
Birth Defects Res B Dev Reprod Toxicol ; 104(3): 117-28, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26195315

RESUMEN

Tabalumab is a human immunoglobulin G subclass 4 monoclonal antibody that has been under development for autoimmune disorders. Tabalumab has full neutralizing activity against both soluble and membrane B-cell activating factor, a B-cell survival factor. The objectives of these studies were to assess the effects of tabalumab on embryo-fetal development and on male (M) and female (F) fertility in rabbits, a pharmacologically relevant species. Doses were administered at 0 (vehicle control), 0.3 (embryo-fetal study only), 1.0, and 30 mg/kg. In the embryo-fetal study, pregnant rabbits does were given a single dose by intravenous injection on gestation day (GD) 7. In the fertility studies, tabalumab was administered by intravenous injection every 7 days starting 2 (F) or 4 (M) weeks before mating, during cohabitation, and until necropsy (M) or through GD 18 (F). Treated animals were mated with untreated partners. Parental clinical signs, body weight, food consumption, blood lymphocyte phenotyping, organ weights, morphologic pathology, ovarian and uterine observations, sperm parameters, and fertility indices were evaluated along with conceptus viability, weight, and morphology. Exposure assessments were made in all main study animals and satellite animals. No adverse parental, reproductive, or developmental effects were observed in any study at any dose. A pharmacodynamic response consisting of dose-dependent decreases in the percent and number of total B lymphocytes and increases in the percent and/or number of total T lymphocytes was observed in parental rabbits at 1.0 and 30 mg/kg. In conclusion, no adverse reproductive or developmental effects were observed in rabbits following exposure to tabalumab at doses as high as 30 mg/kg and exposures at least 14-fold greater than human exposure levels.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Anticuerpos Monoclonales/toxicidad , Fertilidad/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Inmunoglobulina G/toxicidad , Pruebas de Toxicidad , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacocinética , Peso Corporal/efectos de los fármacos , Femenino , Peso Fetal/efectos de los fármacos , Feto/efectos de los fármacos , Feto/embriología , Humanos , Inmunofenotipificación , Linfocitos/efectos de los fármacos , Masculino , Embarazo , Conejos , Reproducción/efectos de los fármacos , Espermatozoides/efectos de los fármacos
16.
Brain Res ; 1583: 237-44, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25128605

RESUMEN

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system and is considered to be caused by the binding of NMO-IgG to aquaporin 4 (AQP4) on astrocytes, which initiates complement-dependent cytotoxicity. AQP4 has two isoforms, i.e., M1 and M23. AQP4 is considered to form heterotetramers containing both isoforms in vivo. Most of the previous studies were performed using either one of the isoforms expressing cell lines. In this study, we generated a fluorescent epitope-tagged AQP4 M1 and M23 co-expressing astrocyte cell line and examined the subcellular targeting of AQP4. In this cell line, AQP4 was targeted mostly to membrane lipid rafts fraction evidenced by sucrose density gradient ultracentrifugation followed by Western blotting with anti-AQP4 antibody. Cholesterol depletion with methyl-ß-cyclodextrin or simvastatin resulted in the dislocation (relocation) of AQP4 from lipid rafts to non-rafts fraction of the membrane and AQP4 was not internalized intracellularly. This change in the localization of AQP4 on membrane significantly reduced complement-dependent cytotoxic effects of NMO-IgG obtained from patients with NMO without affecting AQP4 orthogonal arrays. Thus, these data strongly suggest that the targeting of AQP4 in the lipid rafts is closely related to the pathogenic effects of NMO-IgG.


Asunto(s)
Acuaporina 4/metabolismo , Microdominios de Membrana/metabolismo , Animales , Anticolesterolemiantes/farmacología , Acuaporina 4/genética , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Western Blotting , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Proteína Ácida Fibrilar de la Glía , Humanos , Inmunoglobulina G/toxicidad , Inmunohistoquímica , Isomerismo , Microdominios de Membrana/efectos de los fármacos , Ratones , Microscopía Fluorescente , Proteínas del Tejido Nervioso/metabolismo , Neuromielitis Óptica/inmunología , Simvastatina/farmacología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Transfección , beta-Ciclodextrinas/farmacología
17.
J Neuroimmunol ; 272(1-2): 10-5, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24837568

RESUMEN

This study compares the effects of human antiphospholipid (aPL) and anti-P-ribosomal (anti-P) IgG and control IgG on the brain. Intracerebroventricular (ICV) injected aPL mice (exAPS) displayed specific hyperactivity compared to anti-P-injected (exSLE) and control mice. In contrast ICV injected anti-P-injected mice specifically displayed depression-like behavior and olfactory impairment compared to the other 2 groups. Both anti-P and aPL injected mice were impaired in the passive avoidance test compared to controls. The distinct cognitive effects of the 2 pathogenic antibodies argue for a specific and differential direct action of these autoantibodies on the brain in clinical disease.


Asunto(s)
Anticuerpos Antifosfolípidos/toxicidad , Depresión/inducido químicamente , Hipercinesia/inducido químicamente , Inmunoglobulina G/toxicidad , Trastornos del Olfato/inducido químicamente , Proteínas Ribosómicas/inmunología , Animales , Síndrome Antifosfolípido/sangre , Síndrome Antifosfolípido/inmunología , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Depresión/inmunología , Depresión/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Hipercinesia/inmunología , Inyecciones Intraventriculares , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Trastornos del Olfato/inmunología , Desempeño Psicomotor/efectos de los fármacos , Umbral Sensorial/efectos de los fármacos , Olfato/efectos de los fármacos , Olfato/inmunología
18.
Acta Neuropathol Commun ; 2: 35, 2014 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-24685353

RESUMEN

INTRODUCTION: Antibodies against myelin oligodendrocyte glycoprotein (MOG-IgG) are present in some neuromyelitis optica patients who lack antibodies against aquaporin-4 (AQP4-IgG). The effects of neuromyelitis optica MOG-IgG in the central nervous system have not been investigated in vivo. We microinjected MOG-IgG, obtained from patients with neuromyelitis optica, into mouse brains and compared the results with AQP4-IgG. RESULTS: MOG-IgG caused myelin changes and altered the expression of axonal proteins that are essential for action potential firing, but did not produce inflammation, axonal loss, neuronal or astrocyte death. These changes were independent of complement and recovered within two weeks. By contrast, AQP4-IgG produced complement-mediated myelin loss, neuronal and astrocyte death with limited recovery at two weeks. CONCLUSIONS: These differences mirror the better outcomes for MOG-IgG compared with AQP4-IgG patients and raise the possibility that MOG-IgG contributes to pathology in some neuromyelitis optica patients.


Asunto(s)
Lesiones Encefálicas/inducido químicamente , Inmunoglobulina G/toxicidad , Glicoproteína Mielina-Oligodendrócito/inmunología , Neuromielitis Óptica/inmunología , Animales , Acuaporina 4/inmunología , Astrocitos/efectos de los fármacos , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Moléculas de Adhesión Celular Neuronal/metabolismo , Humanos , Ratones , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuromielitis Óptica/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Canales de Sodio/metabolismo , Factores de Tiempo
19.
J Autoimmun ; 53: 67-77, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24698947

RESUMEN

Spinal cord pathology with inflammatory, demyelinating lesions spanning three or more vertebral segments is a characteristic feature of neuromyelitis optica (NMO). NMO pathogenesis is thought to involve binding of immunoglobulin G anti-aquaporin-4 autoantibodies (NMO-IgG) to astrocytes, causing complement-dependent cytotoxicity (CDC) and secondary inflammation, demyelination and neuron loss. We investigated the involvement of CD59, a glycophosphoinositol (GPI)-anchored membrane protein on astrocytes that inhibits formation of the terminal C5b-9 membrane attack complex. CD59 inhibition by a neutralizing monoclonal antibody greatly increased NMO-IgG-dependent CDC in murine astrocyte cultures and ex vivo spinal cord slice cultures. Greatly increased NMO pathology was also found in spinal cord slice cultures from CD59 knockout mice, and in vivo following intracerebral injection of NMO-IgG and human complement. Intrathecal injection (at L5-L6) of small amounts of NMO-IgG and human complement in CD59-deficient mice produced robust, longitudinally extensive white matter lesions in lumbar spinal cord. Pathology was most severe at day 2 after injection, showing loss of AQP4 and GFAP, C5b-9 deposition, microglial activation, granulocyte infiltration, and demyelination. Hind limb motor function was remarkably impaired as well. There was partial remyelination and recovery of motor function by day 5. Our results implicate CD59 as an important modulator of the immune response in NMO, and provide a novel animal model of NMO that closely recapitulates human NMO pathology. Up-regulation of CD59 on astrocytes may have therapeutic benefit in NMO.


Asunto(s)
Anemia Hemolítica/inmunología , Antígenos CD59/inmunología , Hemoglobinuria/inmunología , Inmunoglobulina G/toxicidad , Neuromielitis Óptica/inmunología , Médula Espinal/inmunología , Anemia Hemolítica/genética , Anemia Hemolítica/patología , Animales , Acuaporina 4/genética , Acuaporina 4/inmunología , Astrocitos/inmunología , Astrocitos/patología , Antígenos CD59/genética , Células Cultivadas , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía , Hemoglobinuria/genética , Hemoglobinuria/patología , Humanos , Inmunoglobulina G/inmunología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Neuromielitis Óptica/inducido químicamente , Neuromielitis Óptica/genética , Neuromielitis Óptica/patología , Médula Espinal/patología
20.
Pain ; 155(2): 299-308, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24145209

RESUMEN

The aetiology of complex regional pain syndrome (CRPS), a highly painful, usually post-traumatic condition affecting the limbs, is unknown, but recent results have suggested an autoimmune contribution. To confirm a role for pathogenic autoantibodies, we established a passive-transfer trauma model. Prior to undergoing incision of hind limb plantar skin and muscle, mice were injected either with serum IgG obtained from chronic CRPS patients or matched healthy volunteers, or with saline. Unilateral hind limb plantar skin and muscle incision was performed to induce typical, mild tissue injury. Mechanical hyperalgesia, paw swelling, heat and cold sensitivity, weight-bearing ability, locomotor activity, motor coordination, paw temperature, and body weight were investigated for 8days. After sacrifice, proinflammatory sensory neuropeptides and cytokines were measured in paw tissues. CRPS patient IgG treatment significantly increased hind limb mechanical hyperalgesia and oedema in the incised paw compared with IgG from healthy subjects or saline. Plantar incision induced a remarkable elevation of substance P immunoreactivity on day 8, which was significantly increased by CRPS-IgG. In this IgG-transfer-trauma model for CRPS, serum IgG from chronic CRPS patients induced clinical and laboratory features resembling the human disease. These results support the hypothesis that autoantibodies may contribute to the pathophysiology of CRPS, and that autoantibody-removing therapies may be effective treatments for long-standing CRPS.


Asunto(s)
Síndromes de Dolor Regional Complejo/inducido químicamente , Síndromes de Dolor Regional Complejo/patología , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Hiperalgesia/patología , Inmunoglobulina G/toxicidad , Adulto , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Síndromes de Dolor Regional Complejo/sangre , Femenino , Humanos , Hiperalgesia/sangre , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Dimensión del Dolor/métodos , Proyectos Piloto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...