Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 697: 149498, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38262291

RESUMEN

Regulatory T cells (Tregs) are lymphocytes that play a central role in peripheral immune tolerance. Tregs are promising targets for the prevention and suppression of autoimmune diseases, allergies, and graft-versus-host disease, and treatments aimed at regulating their functions are being developed. In this study, we created a new modality consisting of a protein molecule that suppressed excessive immune responses by effectively and preferentially expanding Tregs. Recent studies reported that tumor necrosis factor receptor type 2 (TNFR2) expressed on Tregs is involved in the proliferation and activation of Tregs. Therefore, we created a functional immunocytokine, named TNFR2-ICK-Ig, consisting of a fusion protein of an anti-TNFR2 single-chain Fv (scFv) and a TNFR2 agonist TNF-α mutant protein, as a new modality that strongly enhances TNFR2 signaling. The formation of agonist-receptor multimerization (TNFR2 cluster) is effective for the induction of a strong TNFR2 signal, similar to the TNFR2 signaling mechanism exhibited by membrane-bound TNF. TNFR2-ICK-Ig improved the TNFR2 signaling activity and promoted TNFR2 cluster formation compared to a TNFR2 agonist TNF-α mutant protein that did not have an immunocytokine structure. Furthermore, the Treg expansion efficiency was enhanced. TNFR2-ICK-Ig promotes its effects via scFv, which crosslinks receptors whereas the agonists transmit stimulatory signals. Therefore, this novel molecule expands Tregs via strong TNFR2 signaling by the formation of TNFR2 clustering.


Asunto(s)
Anticuerpos de Cadena Única , Linfocitos T Reguladores , Proteínas Portadoras/metabolismo , Proteínas Mutantes/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/agonistas , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Humanos , Animales , Ratones
2.
J Immunol ; 206(8): 1740-1751, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33782090

RESUMEN

Regulatory T cells (Tregs) are a subpopulation of lymphocytes that play a role in suppressing and regulating immune responses. Recently, it was suggested that controlling the functions and activities of Tregs might be applicable to the treatment of human diseases such as autoimmune diseases, organ transplant rejection, and graft-versus-host disease. TNF receptor type 2 (TNFR2) is a target molecule that modulates Treg functions. In this study, we investigated the role of TNFR2 signaling in the differentiation and activation of mouse Tregs. We previously reported the generation of a TNFR2-selective agonist TNF mutant, termed R2agoTNF, by using our unique cytokine modification method based on phage display. R2agoTNF activates cell signaling via mouse TNFR2. In this study, we evaluated the efficacy of R2agoTNF for the proliferation and activation of Tregs in mice. R2agoTNF expanded and activated mouse CD4+CD25+ Tregs ex vivo. The structural optimization of R2agoTNF by internal cross-linking or IgG-Fc fusion selectively and effectively enhanced Treg expansion in vivo. Furthermore, the IgG-Fc fusion protein suppressed skin-contact hypersensitivity reactions in mice. TNFR2 agonists are expected to be new Treg expanders.


Asunto(s)
Enfermedades Autoinmunes , Enfermedad Injerto contra Huésped , Animales , Humanos , Ratones , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Linfocitos T Reguladores , Factor de Necrosis Tumoral alfa
3.
J Biol Chem ; 295(28): 9379-9391, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32398258

RESUMEN

Excessive activation of the proinflammatory cytokine tumor necrosis factor-α (TNFα) is a major cause of autoimmune diseases, including rheumatoid arthritis. TNFα induces immune responses via TNF receptor 1 (TNFR1) and TNFR2. Signaling via TNFR1 induces proinflammatory responses, whereas TNFR2 signaling is suggested to suppress the pathophysiology of inflammatory diseases. Therefore, selective inhibition of TNFR1 signaling and preservation of TNFR2 signaling activities may be beneficial for managing autoimmune diseases. To this end, we developed a TNFR1-selective, antagonistic TNFα mutant (R1antTNF). Here, we developed an R1antTNF derivative, scR1antTNF-Fc, which represents a single-chain form of trimeric R1antTNF with a human IgG-Fc domain. scR1antTNF-Fc had properties similar to those of R1antTNF, including TNFR1-selective binding avidity, TNFR1 antagonistic activity, and thermal stability, and had a significantly extended plasma t1/2in vivo In a murine rheumatoid arthritis model, scR1antTNF-Fc and 40-kDa PEG-scR1antTNF (a previously reported PEGylated form) delayed the onset of collagen-induced arthritis, suppressed arthritis progression in mice, and required a reduced frequency of administration. Interestingly, with these biologic treatments, we observed an increased ratio of regulatory T cells to conventional T cells in lymph nodes compared with etanercept, a commonly used TNF inhibitor. Therefore, scR1antTNF-Fc and 40-kDa PEG-scR1antTNF indirectly induced immunosuppression. These results suggest that selective TNFR1 inhibition benefits the management of autoimmune diseases and that R1antTNF derivatives hold promise as new-modality TNF-regulating biologics.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulina G/farmacología , Mutación Missense , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Sustitución de Aminoácidos , Animales , Línea Celular , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Ratones , Ratones Endogámicos BALB C , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Proteínas Recombinantes de Fusión/genética , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/genética
4.
J Biol Chem ; 292(16): 6438-6451, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28235800

RESUMEN

Tumor necrosis factor-α (TNF) exerts its biological effect through two types of receptors, p55 TNF receptor (TNFR1) and p75 TNF receptor (TNFR2). An inflammatory response is known to be induced mainly by TNFR1, whereas an anti-inflammatory reaction is thought to be mediated by TNFR2 in some autoimmune diseases. We have been investigating the use of an antagonistic TNF mutant (TNFR1-selective antagonistic TNF mutant (R1antTNF)) to reveal the pharmacological effect of TNFR1-selective inhibition as a new therapeutic modality. Here, we aimed to further improve and optimize the activity and behavior of this mutant protein both in vitro and in vivo Specifically, we examined a trimeric structural fusion of R1antTNF, formed via the introduction of short peptide linkers, as a strategy to enhance bioactivity and molecular stability. By comparative analysis with R1antTNF, the trimeric fusion, referred to as single-chain R1antTNF (scR1antTNF), was found to retain in vitro molecular properties of receptor selectivity and antagonistic activity but displayed a marked increase in thermal stability. The residence time of scR1antTNF in vivo was also significantly prolonged. Furthermore, molecular modification using polyethylene glycol (PEG) was easily controlled by limiting the number of reactive sites. Taken together, our findings show that scR1antTNF displays enhanced molecular stability while maintaining biological activity compared with R1antTNF.


Asunto(s)
Proteínas Mutantes/química , Mutación , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/genética , Animales , Antiinflamatorios/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Sitios de Unión , Rastreo Diferencial de Calorimetría , Línea Celular Tumoral , Citocinas/metabolismo , Diseño de Fármacos , Femenino , Fibroblastos/metabolismo , Humanos , Inflamación , Ratones , Ratones Endogámicos BALB C , Polietilenglicoles/química , Conformación Proteica , Ingeniería de Proteínas , Multimerización de Proteína , Receptores Tipo II del Factor de Necrosis Tumoral/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química
5.
J Neurosci ; 36(2): 590-606, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758847

RESUMEN

The synaptic cleft is the space through which neurotransmitters convey neural information between two synaptic terminals. This space is presumably filled with extracellular matrix molecules involved in synaptic function or differentiation. However, little is known about the identities of the matrix components, and it remains unclear how these molecules organize the matrix in synaptic clefts. In this study, we identified Hasp, a Drosophila secretory protein containing CCP and WAP domains. Molecular genetic analysis revealed that Hasp diffuses extracellularly and is predominantly captured at synaptic clefts of cholinergic synapses. Furthermore, Hasp regulates levels of DLG and the nAChR subunits Dα6 and Dα7 at postsynaptic terminals. Hasp is required for trapping of another matrix protein, Hig, which is also secreted and diffused in the brain, at synaptic clefts of cholinergic synapses; however, Hig is dispensable for localization of Hasp at synaptic clefts. In addition, in the brains of triple mutants for the nAChR subunits Dα5, Dα6, and Dα7, the level of Hig, but not Hasp, was markedly reduced in synaptic regions, indicating that these nAChR subunits are required to anchor Hig to synaptic clefts. High-resolution microscopy revealed that Hasp and Hig exhibit segregated distribution within individual synaptic clefts, reflecting their differing roles in synaptogenesis. These data provide insight into how Hasp and Hig construct the synaptic cleft matrix and regulate the differentiation of cholinergic synapses, and also illuminate a previously unidentified architecture within synaptic clefts. SIGNIFICANCE STATEMENT: The synapse has been extensively studied because it is essential for neurotransmission. By contrast, the space between the synaptic terminals, the synaptic cleft, is still an undeveloped research area despite its ubiquity in synapses. In fruit fly brains, we obtained evidence that the matrix protein Hasp and the previously identified Hig, both of which are secreted extracellularly, localize predominantly to synaptic clefts of cholinergic synapses, and modulate the levels of nAChR subunits on postsynaptic membranes. However, Hasp and Hig play differential roles in matrix formation and exhibit segregated distribution within synaptic clefts. These results reveal the molecular mechanisms of synaptic matrix construction and illuminate a molecular architecture within synaptic clefts previously unrevealed in any animal species.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neuronas/ultraestructura , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Drosophila , Proteínas de Drosophila/genética , Regulación de la Expresión Génica/genética , Masculino , Proteínas del Tejido Nervioso/genética , Neurogénesis/fisiología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismo
6.
J Cell Sci ; 128(4): 656-69, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25609706

RESUMEN

Tumor necrosis factor (TNF) is an important mediator that triggers onset of autoimmune diseases and exerts its biological effects by interacting through two receptors, TNFR1 (also known as TNFRSF1A) and TNFR2 (also known as TNFRSF1B). TNFR2 signaling has significant potential to exert pro-survival and protective roles in several diseases. Unlike TNFR1 signaling, however, the mechanism of TNFR2 signal transduction is poorly understood, and few of its adaptor molecules are known. The present study utilized a proteomics approach to search for adaptor molecules in the TNFR2 signaling complex and identified aminopeptidase P3 (APP3, also known as XPNPEP3) to be a key molecule. One of its two isoforms, mitochondrial APP3 (APP3m) but not cytosolic APP3 (APP3c), was recruited to TNFR2 and shown to regulate TNF-TNFR2-dependent phosphorylation of JNK1 (also known as MAPK8) and JNK2 (also known as MAPK9). Furthermore, APP3m was released from mitochondria upon TNF stimulation in the absence of mitochondrial outer membrane permeabilization (MOMP). The observation of increased cell death upon downregulation of APP3m also suggested that APP3m exerts an anti-apoptotic function. These findings reveal that APP3m is a new member of the TNF-TNFR2 signaling complex and characterize an APP3-mediated TNFR2 signal transduction mechanism that induces activation of JNK1 and JNK2.


Asunto(s)
Aminopeptidasas/metabolismo , Mitocondrias/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Apoptosis/fisiología , Transporte Biológico/fisiología , Línea Celular , Células HEK293 , Humanos , Membranas Mitocondriales/metabolismo , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal
7.
Nanotechnology ; 28(13): 135101, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28240988

RESUMEN

Many of the beneficial and toxic biological effects of nanoparticles have been shown to have a negative correlation with particle size. However, few studies have demonstrated biological effects that only occur at specific nanoparticle sizes. Further elucidation of the size-specific biological effects of nanoparticles may reveal not only unknown toxicities, but also novel benefits of nanoparticles. We used surface-unmodified silica particles with a wide range of diameters and narrow size intervals between the diameters (10, 30, 50, 70, 100, 300, and 1000 nm) to investigate the relationship between particle size and acute toxicity after intravenous administration in mice. Negative correlations between particle size and thrombocytopenia, liver damage, and lethal toxicity were observed. However, a specific size-effect was observed for the severity of hypothermia, where silica nanoparticles with a diameter of 50 nm induced the most severe hypothermia. Further investigation revealed that this hypothermia was mediated not by histamine, but by platelet-activating factor, and it was independent of the thrombocytopenia and the liver damage. In addition, macrophages/Kupffer cells and platelets, but not neutrophils, play a critical role in the hypothermia. The present results reveal that silica nanoparticles have particle size-specific toxicity in mice, suggesting that other types of nanoparticles may also have biological effects that only manifest at specific particle sizes. Further study of the size-specific effects of nanoparticles is essential for safer and more effective nanomedicines.


Asunto(s)
Nanopartículas/administración & dosificación , Nanopartículas/química , Tamaño de la Partícula , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/química , Administración Intravenosa , Animales , Plaquetas/metabolismo , Femenino , Hipotermia Inducida , Macrófagos del Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Nanopartículas/toxicidad , Factor de Activación Plaquetaria/metabolismo , Dióxido de Silicio/toxicidad , Pruebas de Toxicidad Aguda
8.
Biol Pharm Bull ; 40(5): 726-728, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458361

RESUMEN

We previously reported that unmodified silica nanoparticles with diameters of 70 nm (nSP70) induced liver damage in mice, whereas nSP70 modified with carboxyl or amino groups did not. In addition, we have found that both unmodified and modified nSP70s localize in both Kupffer cells and parenchymal hepatocytes. We therefore evaluated the contributions of nSP70 uptake by these cell populations to liver damage. To this end, we pretreated mice with gadolinium (III) chloride hydrate (GdCl3) to prevent nSP70 uptake by Kupffer cells, subsequently injected the mice with either type of nSP70, and then assessed plasma levels of alanine aminotransferase (ALT). In mice given GdCl3, unmodified nSP70 increased ALT levels. From these data, we hypothesized that in GdCl3-treated mice, the unmodified nSP70 that was prevented from entering Kupffer cells was shunted to parenchymal hepatocytes, where it induced cytotoxicity and increased liver damage. In contrast, GdCl3 pretreatment had no effect on ALT levels in mice injected with surface-modified nSP70s, suggesting that modified nSP70s spared parenchymal hepatocytes and thus induced negligible liver damage. In cytotoxicity analyses, the viability of a parenchymal hepatocyte line was greater when exposed to surface-modified nSP70s than to unmodified nSP70s. These findings imply that the decreased liver damage associated with surface-modified compared with unmodified nSP70 is attributable to decreased cytotoxicity to parenchymal hepatocytes.


Asunto(s)
Aminas/química , Ácidos Carboxílicos/química , Nanopartículas/química , Dióxido de Silicio/química , Alanina Transaminasa/análisis , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Femenino , Gadolinio/química , Hepatocitos/efectos de los fármacos , Macrófagos del Hígado/efectos de los fármacos , Pruebas de Función Hepática , Ratones , Ratones Endogámicos BALB C , Nanopartículas/toxicidad , Tamaño de la Partícula , Dióxido de Silicio/toxicidad , Propiedades de Superficie
9.
Am J Physiol Heart Circ Physiol ; 311(2): H476-86, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27342875

RESUMEN

Acute myocarditis is a self-limiting disease. Most patients with myocarditis recover without cardiac dysfunction in spite of limited capacity of myocardial regeneration. Therefore, to address intrinsic reparative machinery of inflamed hearts, we investigated the cellular dynamics of cardiomyocytes in response to inflammation using experimental autoimmune myocarditis (EAM) model. EAM was induced by immunization of BALB/c mice with α-myosin heavy chain peptides twice. The inflammatory reaction was evoked with myocardial damage with the peak at 3 wk after the first immunization (EAM3w). Morphological and functional restoration started from EAM3w, when active protrusion formation, a critical process of myocardial healing, was observed in cardiomyocytes. Shotgun proteomics revealed that cytoskeletal proteins were preferentially increased in cardiomyocytes at EAM3w, compared with preimmunized (EAM0w) hearts, and that moesin was the most remarkably upregulated among them. Immunoblot analyses demonstrated that the expression of both total and phosphorylated moesin was upregulated in isolated cardiomyocytes from EAM3w hearts. Immunofluorescence staining showed that moesin was localized at cardiomyocyte protrusions at EAM3w. Adenoviral vectors expressing wild-type, constitutively active and inactive form of moesin (wtMoesin, caMoesin, and iaMoesin, respectively) were transfected in neonatal rat cardiomyocytes. The overexpression of wtMoesin and caMoesin resulted in protrusion formation, while not iaMoesin. Finally, we found that cardiomyocyte protrusions were accompanied by cell-cell contact formation. The expression of moesin was upregulated in cardiomyocytes under inflammation, inducing protrusion formation in a phosphorylation-dependent fashion. Moesin signal could be a novel therapeutic target that stimulates myocardial repair by promoting contact formation of cardiomyocytes.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Extensiones de la Superficie Celular/genética , Citoesqueleto/metabolismo , Inflamación/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocarditis/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Enfermedades Autoinmunes/inducido químicamente , Extensiones de la Superficie Celular/patología , Supervivencia Celular , Citoesqueleto/patología , Modelos Animales de Enfermedad , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Miocarditis/inducido químicamente , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/efectos adversos , Péptidos , Fosfoproteínas/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Chembiochem ; 17(2): 181-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26561285

RESUMEN

Hypoxia-adapted cancer cells in tumors contribute to the pathological progression of cancer. Cancer research has therefore focused on the identification of molecules responsible for hypoxia adaptation in cancer cells, as well as the development of new compounds with action against hypoxia-adapted cancer cells. The marine natural product furospinosulin-1 (1) has displayed hypoxia-selective growth inhibition against cultured cancer cells, and has shown in vivo anti-tumor activity, although its precise mode of action and molecular targets remain unclear. In this study, we found that 1 is selectively effective against hypoxic regions of tumors, and that it directly binds to the transcriptional regulators p54(nrb) and LEDGF/p75, which have not been previously identified as mediators of hypoxia adaptation in cancer cells.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/química , Proteínas Asociadas a Matriz Nuclear/química , Factores de Transcripción de Octámeros/química , Proteínas de Unión al ARN/química , Sesterterpenos/química , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Hipoxia de la Célula/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Proteínas de Unión al ADN , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Estructura Molecular , Neoplasias/tratamiento farmacológico , Unión Proteica/efectos de los fármacos , Sesterterpenos/farmacología , Sesterterpenos/uso terapéutico
11.
Bioorg Med Chem Lett ; 26(2): 530-533, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26627581

RESUMEN

Recently, 7-substituted 7-deazapurine nucleoside triphosphates and 5-substituted pyrimidine nucleoside triphosphates (dN(am)TPs) were synthesized to extend enzymatically using commercially available polymerase. However, extension was limited when we attempted to incorporate the substrates consecutively. To address this, we have produced a mutant polymerase that can efficiently accept the modified nucleotide with amphiphilic groups as substrates. Here we show that the KOD polymerase mutant, KOD exo(-)/A485L, had the ability to incorporate dN(am)TP continuously over 50nt, indicating that the mutant is sufficient for generating functional nucleic acid molecules.


Asunto(s)
ADN Polimerasa Dirigida por ADN/química , Oligodesoxirribonucleótidos/química , Nucleótidos de Purina/química , Nucleótidos de Pirimidina/química , ADN Polimerasa Dirigida por ADN/genética , Oligodesoxirribonucleótidos/genética , Mutación Puntual , Polietilenglicoles/química , Nucleótidos de Purina/genética , Nucleótidos de Pirimidina/genética , Temperatura
12.
Biochem Biophys Res Commun ; 456(4): 908-12, 2015 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-25528586

RESUMEN

The EPH receptor A10 (EphA10) is up-regulated in breast cancer but is not normally expressed in healthy tissue, thus it has been suggested that EphA10 may be a useful target for cancer therapy. This study reports a diabody, an antibody derivative binding two different target molecules, EphA10 expressed in tumor cells and CD3 expressed in T cells, which showed T cell dependent-cytotoxicity. The diabody, which has His-tagged and FLAG-tagged chains, was expressed in Escherichia coli and purified in both heterodimer (Db-1) and homodimer (Db-2) formulations by liquid chromatography. Flow cytometry analysis using EphA10-expressing cells showed that binding activity of heterodimers was stronger than that of homodimers. Addition of diabodies to PBMC cultures resulted in T-cell mediated redirected lysis, and the bioactivity was consistent with the stronger binding activity of heterodimeric diabody formulations. Our results indicate that diabodies recognizing both EphA10 and CD3 could have a range of potential applications in cancer therapy, such as breast cancers that express the EPH receptor A10, especially triple negative breast cancer.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Biespecíficos/inmunología , Complejo CD3/inmunología , Receptores de la Familia Eph/inmunología , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Unión Proteica , Transfección
13.
Blood ; 121(14): 2804-13, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23365463

RESUMEN

Monoclonal antibodies (mAbs) that are internalized into cells are a current focus in the development of antibody-drug conjugates (ADCs). We describe a phage display-based high-throughput screening system to rapidly isolate cell-internalizing mAbs. We simultaneously examined the cell-internalizing activities of several hundred independent mAbs and successfully isolated cell-internalizing mAbs against the tumor endothelial markers Roundabout homolog 4 (Robo4) and vascular endothelial growth factor receptor 2 (VEGFR2). Tumor accumulation of mAbs with high cell-internalizing activity was significantly higher than that of mAbs with low cell-internalizing activity. Furthermore, the antitumor effects of ADCs of mAbs with high cell-internalizing activity were significantly stronger than those of mAbs with low cell-internalizing activity. Although anti-VEGFR2 therapy caused a significant loss of body weight, anti-Robo4 therapy did not. These findings indicate that cell-internalizing activity plays an important role in the biodistribution and therapeutic effects of ADCs. Further, Robo4 can be an effective marker for tumor vascular targeting.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Inmunoconjugados/farmacocinética , Melanoma/terapia , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Transporte Biológico/inmunología , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Línea Celular Transformada , Diseño de Fármacos , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunoconjugados/inmunología , Melanoma/irrigación sanguínea , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Neovascularización Patológica/terapia , Biblioteca de Péptidos , Receptores de Superficie Celular , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inmunología , Distribución Tisular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Bioorg Med Chem Lett ; 25(15): 2888-91, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26048797

RESUMEN

Here, we describe the enzymatic construction of a new larger base pair formed between adenine (A) and a 4-hydroxy-2-mercaptobenzimidazole (SB) nucleobase analogue. We investigated the enzymatic incorporation of 2'-deoxynucleoside-5'-triphosphate bearing a SB nucleobase analogue (dSBTP) into oligonucleotides (ONs) by DNA polymerases. dSBTP could be effectively incorporated at the site opposite a dA in a DNA template by several B family DNA polymerases. These findings provide new insights into various aspects of biotechnology, including the design of non-natural base pairs.


Asunto(s)
Adenina/metabolismo , Bencimidazoles/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Nucleótidos/metabolismo , Polifosfatos/metabolismo , Adenina/química , Emparejamiento Base , Secuencia de Bases , Bencimidazoles/química , Cartilla de ADN/química , Cartilla de ADN/metabolismo , Nucleótidos/química , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Polimerizacion , Polifosfatos/química
15.
Nanotechnology ; 26(24): 245101, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-26011124

RESUMEN

Recently, nanomaterial-mediated biological effects have been shown to be governed by the interaction of nanomaterials with some kinds of proteins in biological fluids, and the physical characteristics of the nanomaterials determine the extent and type of their interactions with proteins. Here, we examined the relationships between the surface properties of amorphous silica nanoparticles with diameters of 70 nm (nSP70), their interactions with some proteins in biological fluids, and their toxicity in mice after intravenous administration. The surface modification of nSP70 with amino groups (nSP70-N) prevented acute lethality and abnormal activation of the coagulation cascade found in the nSP70-treated group of mice. Since our previous study showed that coagulation factor XII played a role in the nSP70-mediated abnormal activation of the coagulation cascade, we examined the interaction of nSP70 and nSP70-N with coagulation factor XII. Coagulation factor XII bonded to the surface of nSP70 to a greater extent than that observed for nSP70-N, and consequently more activation of coagulation factor XII was observed for nSP70 than for nSP70-N. Collectively, our results suggest that controlling the interaction of nSP70 with blood coagulation factor XII by modifying the surface properties would help to inhibit the nSP70-mediated abnormal activation of the blood coagulation cascade.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Nanopartículas/toxicidad , Corona de Proteínas/metabolismo , Dióxido de Silicio/toxicidad , Administración Intravenosa , Animales , Factor XIIa/metabolismo , Femenino , Ratones , Nanopartículas/administración & dosificación , Tamaño de la Partícula , Dióxido de Silicio/administración & dosificación , Propiedades de Superficie
16.
Part Fibre Toxicol ; 12: 16, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-26113229

RESUMEN

BACKGROUND: The skin is a key route of human exposure to nanomaterials, which typically occurs simultaneously with exposure to other chemical and environmental allergen. However, little is known about the hazards of nanomaterial exposure via the skin, particularly when accompanied by exposure to other substances. RESULTS: Repeated topical treatment of both ears and the shaved upper back of NC/Nga mice, which are models for human atopic dermatitis (AD), with a mixture of mite extract and silica nanoparticles induced AD-like skin lesions. Measurements of ear thickness and histologic analyses revealed that cutaneous exposure to silica nanoparticles did not aggravate AD-like skin lesions. Instead, concurrent cutaneous exposure to mite allergens and silica nanoparticles resulted in the low-level production of allergen-specific IgGs, including both the Th2-related IgG1 and Th1-related IgG2a subtypes, with few changes in allergen-specific IgE concentrations and in Th1 and Th2 immune responses. In addition, these changes in immune responses increased the sensitivity to anaphylaxis. Low-level IgG production was induced when the mice were exposed to allergen-silica nanoparticle agglomerates but not when the mice exposed to nanoparticles applied separately from the allergen or to well-dispersed nanoparticles. CONCLUSIONS: Our data suggest that silica nanoparticles themselves do not directly affect the allergen-specific immune response after concurrent topical application of nanoparticles and allergen. However, when present in allergen-adsorbed agglomerates, silica nanoparticles led to a low IgG/IgE ratio, a key risk factor of human atopic allergies. We suggest that minimizing interactions between nanomaterials and allergens will increase the safety of nanomaterials applied to skin.


Asunto(s)
Anafilaxia/inmunología , Antígenos Dermatofagoides , Dermatitis Alérgica por Contacto/inmunología , Inmunoglobulina E/inmunología , Nanopartículas , Dióxido de Silicio , Piel/inmunología , Anafilaxia/sangre , Animales , Citocinas/sangre , Citocinas/inmunología , Dermatitis Alérgica por Contacto/sangre , Dermatitis Alérgica por Contacto/patología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Medición de Riesgo , Piel/patología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Factores de Tiempo
17.
Biochem Biophys Res Commun ; 450(1): 545-9, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24924629

RESUMEN

We recently identified Eph receptor A10 (EphA10) as a novel breast cancer-specific protein. Moreover, we also showed that an in-house developed anti-EphA10 monoclonal antibody (mAb) significantly inhibited proliferation of breast cancer cells, suggesting EphA10 as a promising target for breast cancer therapy. However, the only other known report for EphA10 was its expression in the testis at the mRNA level. Therefore, the potency of EphA10 as a drug target against cancers other than the breast is not known. The expression of EphA10 in a wide variety of cancer cells was studied and the potential of EphA10 as a drug target was evaluated. Screening of EphA10 mRNA expression showed that EphA10 was overexpressed in breast cancer cell lines as well as in prostate and colon cancer cell lines. Thus, we focused on prostate cancers in which EphA10 expression was equivalent to that in breast cancers. As a result, EphA10 expression was clearly shown in clinical prostate tumor tissues as well as in cell lines at the mRNA and protein levels. In order to evaluate the potential of EphA10 as a drug target, we analyzed complement-dependent cytotoxicity effects of anti-EphA10 mAb and found that significant cytotoxicity was mediated by the expression of EphA10. Therefore, the idea was conceived that the overexpression of EphA10 in prostate cancers might have a potential as a target for prostate cancer therapy, and formed the basis for the studies reported here.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores de la Familia Eph/metabolismo , Anticuerpos Monoclonales/inmunología , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Masculino , Neoplasias de la Próstata/patología , Receptores de la Familia Eph/inmunología , Resultado del Tratamiento
18.
Protein Sci ; 33(7): e5084, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38923711

RESUMEN

Small antibody fragments have recently been used as alternatives to full-length monoclonal antibodies in therapeutic applications. One of the most popular fragment antibodies is single-chain fragment variables (scFvs), consisting of variable heavy (VH) and variable light (VL) domains linked by a flexible peptide linker. scFvs have small molecular sizes, which enables good tissue penetration and low immunogenicity. Despite these advantages, the use of scFvs, especially for therapeutic purpose, is still limited because of the difficulty to regulate the binding activity and conformational stability. In this study, we constructed and analyzed 10 scFv fragments derived from 10 representatives of FDA-approved mAbs to evaluate their physicochemical properties. Differential scanning calorimetry analysis showed that scFvs exhibited relatively high but varied thermostability, from 50 to 70°C of melting temperatures, and different unfolding cooperativity. Surface plasmon resonance analysis revealed that scFvs fragments that exhibit high stability and cooperative unfolding likely tend to maintain antigen binding. This study demonstrated the comprehensive physicochemical properties of scFvs derived from FDA-approved antibodies, providing insights into antibody design and development.


Asunto(s)
Estabilidad Proteica , Anticuerpos de Cadena Única , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología , Humanos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Rastreo Diferencial de Calorimetría , Unión Proteica
19.
Cytokine ; 61(2): 578-84, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23246116

RESUMEN

The cytokine lymphotoxin-α (LTα) is a promising candidate for use in cancer therapy. However, the instability of LTαin vivo and the insufficient levels of tumor necrosis factor receptor 1 (TNFR1)-mediated bioactivity of LTα limit its therapeutic potential. Here, we created LTα mutants with increased TNFR1-mediated bioactivity by using a phage display technique. We constructed a phage library displaying lysine-deficient structural variants of LTα with randomized amino acid residues. After affinity panning, we screened three clones of lysine-deficient LTα mutant, and identified a LTα mutant with TNFR1-mediated bioactivity that was 32 times that of the wild-type LTα (wtLTα). When compared with wtLTα, the selected clone showed augmented affinity to TNFR1 due to slow dissociation rather than rapid association. In contrast, the mutant showed only 4 times the TNFR2-mediated activity of wtLTα. In addition, the LTα mutant strongly and rapidly activated caspases that induce TNFR1-mediated cell death, whereas the mutant and wtLTα activated nuclear factor-kappa B to a similar extent. Our data suggest that the kinetics of LTα binding to TNFR1 play an important role in signal transduction patterns, and a TNFR1-selective LTα mutant with augmented bioactivity would be a superior candidate for cancer therapy.


Asunto(s)
Linfotoxina-alfa/uso terapéutico , Proteínas Mutantes/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Espectroscopía de Resonancia por Spin del Electrón , Activación Enzimática/efectos de los fármacos , Humanos , Punto Isoeléctrico , Cinética , Linfotoxina-alfa/química , Linfotoxina-alfa/farmacología , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/farmacología , FN-kappa B/metabolismo , Neoplasias/enzimología , Unión Proteica/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/química
20.
Nanotechnology ; 24(40): 405102, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24029385

RESUMEN

Although nanomaterials are being used in various fields, their safety is not yet sufficiently understood. We have been attempting to establish a nanomaterials safety-assessment system by using biomarkers to predict nanomaterial-induced adverse biological effects. Here, we focused on microRNAs (miRNAs) because of their tissue-specific expression and high degree of stability in the blood. We previously showed that high intravenous doses of silica nanoparticles of 70 nm diameter (nSP70) induced liver damage in mice. In this study, we compared the effectiveness of serum levels of liver-specific or -enriched miRNAs (miR-122, miR-192, and miR-194) with that of conventional hepatic biomarkers (alanine aminotransferase (ALT) and aspartate aminotransferase (AST)) as biomarkers for nSP70. After mice had been treated with nSP70, their serum miRNAs levels were measured by using quantitative RT-PCR. Serum levels of miR-122 in nSP70-treated mice were the highest among the three miRNAs. The sensitivity of miR-122 for liver damage was at least as good as those of ALT and AST. Like ALT and AST, miR-122 may be a useful biomarker of nSP70. We believe that these findings will help in the establishment of a nanomaterials safety-assessment system.


Asunto(s)
Biomarcadores/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , MicroARNs/sangre , Nanopartículas/toxicidad , Animales , Femenino , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Sensibilidad y Especificidad , Dióxido de Silicio/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA