Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Ethnopharmacol ; 321: 117544, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38070838

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Modified Danzhi Xiaoyao Powder (MDXP) is a traditional Chinese medicine formula remedy for treating Dry Eye Disease (DED). It showed the function of dispersing stagnated liver Qi for relieving Qi stagnation and clearing heat, which can be effective in treating conditions such as Dry Eye Disease (DED) and irregular menstruation due to liver depression and fire transformation. AIM OF THE STUDY: This study investigated the mechanism of the effect of MDXP in mice with DED. MATERIALS AND METHODS: A DED model was induced in mice using chronic painful stimulation (tail clamping) in combination with Benzalkonium Chloride Solution drops administered in a dry box for 28 days. After modeling, the MDXP groups were given Chinese medicine with different dosages by gavage for 14 days. The following parameters were recorded in each group: body mass, anal temperature, tear secretion, tear film rupture time, and corneal fluorescein staining. Behavioral changes were evaluated by elevating cross-maze and open-field experiments. The levels of inflammatory factors serum tumor necrosis factor-α (TNF-α), interleukin 1ß (IL-1ß), fcγR-mediated phagocytosis pathway cell division control protein 42 homolog (CDC42), actin-related protein 2/3 complex subunit 2 (ARPC2), and actin-related protein 3 (ACTR3) were measured by using Enzyme-linked immunoassay (ELISA), immunohistochemical staining, and real-time fluorescent qualitative polymerase chain reaction (RT-qPCR). RESULTS: MDXP increased body mass and lowered body temperature, prolonged tear film break-up time, promoted tear secretion, repaired corneal damage, decreased horizontal and vertical scores, elevated percentage of open arm times and boom opening time percentage, and reduced the expression levels of inflammatory factors of TNF-α, IL-1ß and pathway-related proteins CDC42, ARPC2, and ACTR3 in mice. MDXP also reduced the expression levels of inflammatory factors of TNF-α and IL-1ß in human corneal endothelial cells (HCECs), mouse mononuclear macrophage cells (RAW264.7), and human myeloid leukemia mononuclear cells (THP-1). CONCLUSIONS: MDXP can relieve tension and anxiety, inhibit apoptosis, reduce phagocytosis, reduce the expression of pro-inflammatory factors, repair corneal damage, and improve the symptoms in DED mice. The mechanism of action may be through the fcγR-mediated phagocytosis pathway.


Asunto(s)
Lesiones de la Cornea , Síndromes de Ojo Seco , Femenino , Humanos , Ratones , Animales , Polvos/uso terapéutico , Factor de Necrosis Tumoral alfa , Células Endoteliales/metabolismo , Receptores de IgG/uso terapéutico , Síndromes de Ojo Seco/tratamiento farmacológico , Fagocitosis
2.
Rev Med Virol ; 33(2): e2422, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36658757

RESUMEN

Dengue fever, the most common arbovirus disease, affects an estimated 390 million people annually. Dengue virus (DENV) is an RNA virus of the Flaviviridae family with four different serotypes. Dengue haemorrhagic fever is the deadliest form of dengue infection and is characterised by thrombocytopaenia, hypotension, and the possibility of multi-system organ failure. The mechanism hypothesised for DENV viral replication is intrinsic antibody-dependent enhancement, which refers to Fcγ receptor-mediated viral amplification. This hypothesis suggests that the internalisation of DENV through the Fcγ receptor inhibits antiviral genes by suppressing type-1 interferon-mediated antiviral responses. DENV NS1 antibodies can promote the release of various inflammatory mediators in the nuclear transcription factor pathway (NF-κB-dependent), including monocyte chemoattractant protein (MCP)-1, interleukin (IL)-6, and IL-8. As a result, MCP-1 increases ICAM-1 expression and facilitates leukocyte transmigration. In addition, anti-DENV NS1 antibodies induce endothelial cell apoptosis via a nitric oxide-regulated pathway. A chain reaction involving pre-existing DENV heterotypic antibodies and innate immune cells causes dysfunction in complement system activity and contributes to the action of autoantibodies and anti-endothelial cells, resulting in endothelial cell dysfunction, blood-retinal barrier breakdown, haemorrhage, and plasma leakage. A spectrum of ocular diseases associated with DENV infection, ranging from haemorrhagic to inflammatory manifestations, has been reported in the literature. Although rare, ophthalmic manifestations can occur in both the anterior and posterior segments and are usually associated with thrombocytopenia. The most common ocular complication is haemorrhage. However, ophthalmic complications, such as anterior uveitis and vasculitis, suggest an immune-mediated pathogenesis.


Asunto(s)
Virus del Dengue , Dengue , Trombocitopenia , Humanos , Receptores de IgG/uso terapéutico , Hemorragia/complicaciones , Interleucina-6 , Antivirales/uso terapéutico
3.
Expert Opin Investig Drugs ; 31(7): 715-727, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35531637

RESUMEN

INTRODUCTION: Primary immune thrombocytopenia (ITP) is an acquired autoimmune disease of unknown cause. Autoantibodies, self-reactive T cells and other immune abnormalities, with impairment of platelet production, lead to a reduced platelet count. Until recently, therapy was largely empirical using immune suppressants (none of which have undergone randomized clinical trials). These therapies have variable efficacy and are associated with predictable unwanted effects which impact patient quality-of-life. With greater understanding of the underlying pathophysiology, better, more targeted therapies have been developed; however, there is still an urgent need for additional classes of treatment. AREAS COVERED: This article covers new TPO receptor agonists, Syk inhibitors, Fcγ receptor antagonists, BTK and complement inhibitors, and other therapies. Insights into the most promising therapies are offered. Novel ITP treatments currently in clinical trials and those recently approved come under the spotlight. EXPERT OPINION: Thrombopoietin receptor agonists remain the most effective treatment for ITP and have changed the ITP therapeutic landscape remarkably. Other new molecules such as Fcγ receptor blockers, Bruton tyrosine kinase, complement inhibitors, and others are unlikely to enjoy the same success rate as the TPO-RAs, but nonetheless they will find a place in the management of patients with ITP.


Asunto(s)
Púrpura Trombocitopénica Idiopática , Inactivadores del Complemento , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Humanos , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Receptores de IgG/uso terapéutico , Receptores de Trombopoyetina/agonistas , Receptores de Trombopoyetina/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Trombopoyetina/agonistas , Trombopoyetina/uso terapéutico
4.
Sci Rep ; 9(1): 11392, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31388039

RESUMEN

Skeletal muscle is under inhibitory homeostatic regulation by multiple ligands of the transforming growth factor-ß (TGFß) superfamily. Follistatin is a secreted protein that promotes muscle growth and function by sequestering these ligands extracellularly. In the present study, we evaluated the potential of ACE-083 - a locally acting, follistatin-based fusion protein - as a novel therapeutic agent for focal or asymmetric myopathies. Characterization of ACE-083 in vitro revealed its high affinity for heparin and extracellular matrix while surface plasmon resonance and cell-based assays confirmed that ACE-083 binds and potently neutralizes myostatin, activin A, activin B and growth differentiation factor 11 (GDF11). Intramuscular administration of ACE-083 caused localized, dose-dependent hypertrophy of the injected muscle in wild-type mice and mouse models of Charcot-Marie-Tooth disease (CMT) and Duchenne muscular dystrophy, with no evidence of systemic muscle effects or endocrine perturbation. Importantly, ACE-083 also increased the force of isometric contraction in situ by the injected tibialis anterior muscle in wild-type mice and disease models and increased ankle dorsiflexion torque in CMT mice. Our results demonstrate the potential of ACE-083 as a therapeutic agent for patients with CMT, muscular dystrophy and other disorders with focal or asymmetric muscle atrophy or weakness.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/tratamiento farmacológico , Folistatina/farmacología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Activinas/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Modelos Animales de Enfermedad , Folistatina/genética , Folistatina/uso terapéutico , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Hipertrofia/inducido químicamente , Ligandos , Masculino , Ratones , Ratones Endogámicos mdx , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Miostatina/metabolismo , Receptores de IgG/genética , Receptores de IgG/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico
5.
Immunol Rev ; 268(1): 160-74, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26497519

RESUMEN

Fcγ receptor I (FcγRI or CD64) is the sole human Fc receptor with high affinity for monovalent IgG. While it contains an immunoreceptor tyrosine-based activation motif in its cytoplasmic domain, binding of FcγRI can result in a complex array of activating and inhibitory outcomes. For instance, binding of monomeric IgG provides a low-intensity tonic signal through FcγRI that is necessary for full interferon γ receptor signaling in the same cell. Interaction of FcγRI with larger high-avidity complexes can result in phagocytosis, the generation of reactive oxygen species, as well as the synthesis and release of inflammatory cytokines. However, numerous reports also document potent anti-inflammatory effects brought about by FcγRI engagement with immune complexes such as the inhibition of IFNγ and TLR4 signaling, and secretion of interleukin-10. This has led to conflicting hypotheses regarding the function of FcγRI, especially with regard to its role in the efficacy of several therapeutic monoclonal antibodies. While many of these issues are still unclear, continued characterization of the regulation and context dependence of FcγRI function, as well as the molecular mechanisms responsible for these various outcomes, will improve our understanding of FcγRI biology as well as the therapeutic strategies designed to harness or constrain its actions.


Asunto(s)
Inmunoglobulina G/metabolismo , Receptores de IgG/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Regulación de la Expresión Génica , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Inmunoterapia , Infecciones/inmunología , Infecciones/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Interferón gamma/metabolismo , Ligandos , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Especificidad de Órganos/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de IgG/química , Receptores de IgG/genética , Receptores de IgG/uso terapéutico , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
8.
J Immunol ; 189(12): 5513-7, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23150715

RESUMEN

mAb therapy for experimental metastatic melanoma relies on activating receptors for the Fc portion of IgG (FcγR). Opposing results on the respective contribution of mouse FcγRI, FcγRIII, and FcγRIV have been reported using the gp75-expressing B16 melanoma and the protective anti-gp75 mAb TA99. We analyzed the contribution of FcγRs to this therapy model using bioluminescent measurement of lung metastases loads, novel mouse strains, and anti-FcγR blocking mAbs. We found that the TA99 mAb-mediated effects in a combination therapy using cyclophosphamide relied on activating FcγRs. The combination therapy, however, was not more efficient than mAb therapy alone. We demonstrate that FcγRI and, unexpectedly, FcγRIII contributed to TA99 mAb therapeutic effects, whereas FcγRIV did not. Therefore, FcγRIII and FcγRI are, together, responsible for anti-gp75 mAb therapy of B16 lung metastases. Our finding that mouse FcγRIII contributes to Ab-induced tumor reduction correlates with clinical data on its human functional equivalent human FcγRIIIA (CD16A).


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Receptores de IgG/uso terapéutico , Proteínas Virales/inmunología , Animales , Anticuerpos Bloqueadores/uso terapéutico , Arbovirus/inmunología , Hibridomas , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de IgG/deficiencia , Receptores de IgG/genética
9.
Cancer Genomics Proteomics ; 9(6): 357-72, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23162075

RESUMEN

In this review, we summarize approaches to treat cancer with genetically engineered fusion proteins. Such proteins can act as decoy receptors for several ligands or as recruiters of immune effector cells to tumor. Examples of interference with growth factor-mediated tumor growth and tumor-related angiogenesis with fusion proteins consisting of the extracellular domains, and in some cases also of entities of one or several receptors and the Fc part of human IgG1, are discussed. In addition, we present strategies for recruitment of immune effector cells to tumor with fusion proteins. This can be achieved with fusion proteins consisting of a tumor-related antibody and a cytokine or major histocompatibilty complex class-I-peptide complexes, by T-cell receptor cytokine fusion proteins or by combination of a T-cell-recruiting antibody with a tumor-related ligand or a defined T-cell receptor.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Citocinas/uso terapéutico , Humanos , Receptores de Antígenos de Linfocitos T/uso terapéutico , Receptores de IgG/uso terapéutico
10.
Arthritis Res Ther ; 14(4): R192, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22906120

RESUMEN

INTRODUCTION: Soluble immune aggregates bearing intact Fc fragments are effective treatment for a variety of autoimmune disorders in mice. The better to understand the mechanisms by which Fc-bearing immune complexes suppress autoimmunity, and to develop a platform for clinical translation, we created a series of fully recombinant forms of polyvalent IgG2a Fc, termed stradomers, and tested their efficacy in a therapeutic model of collagen-induced arthritis (CIA) and preventive models of both idiopathic thrombocytopenic purpura (ITP) and graft-versus-host disease (GVHD). METHODS: Stradomers were created by engineering either the human IgG2 hinge sequence (IgG2H) or the isoleucine zipper (ILZ) onto either the carboxy or amino termini of murine IgG2a Fc. Multimerization and binding to the canonical Fc receptors and the C-type lectin SIGN-RI were evaluated by using sodium dodecylsulfate-polymerase chain reaction (SDS-PAGE) and Biacore/Octet assays. The efficacy of stradomers in alleviating CIA and preventing ITP and GVHD was compared with "gold standard" therapies, including prednisolone and intravenous immune globulin (IVIG). RESULTS: Stradomers exist as both homodimeric and highly ordered sequential multimers. Higher-order multimers demonstrate increasingly stable associations with the canonic Fcγ receptors (FcγRs), and SIGN-R1, and are more effective than Fc homodimers in treating CIA. Furthermore, stradomers confer partial protection against platelet loss in a murine model ITP, but do not prevent GVHD. CONCLUSION: These data suggest that fully human stradomers might serve as valuable tools for the treatment of selected autoimmune disorders and as reagents to study the function of Fc:FcR interactions in vivo.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Púrpura Trombocitopénica Idiopática/prevención & control , Receptores de IgG/uso terapéutico , Animales , Artritis Experimental/metabolismo , Femenino , Inmunoglobulina G/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Púrpura Trombocitopénica Idiopática/metabolismo , Distribución Aleatoria , Receptores de IgG/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
11.
Rev. Fac. Med. (Caracas) ; 35(1): 40-44, ene.-jun. 2012. tab
Artículo en Español | LILACS | ID: lil-682991

RESUMEN

En estudios previos, se ha descrito un disminución de la activación y actividad citotóxica de las células NK en los pacientes infectados con hepatitis C; sin embargo, se desconoce el mecanismo por el cual éste fenómeno ocurre. En el presente reporte se estudió el efecto de la proteína E2 de la envoltura del virus o de la estimulación de su receptor con el anticuerpo anti-CD81 sobre la fosforilación de tirosinas, serinas, las enzimas: proteína quinasa C y fosfoinositol 3 quinasa, el factor de transcrición Nfkb y el intercambiador de nueclotidos VAV de células NK de controles normales estimulados con anti-CD16. Ambos, la proteína E2 y anti-CD81, combinado o por separado inducen una disminución de la fosforilación de tirosinas y serinas, así como una marcada disminución de la fosforilación de PKC, NFkB, PI3K y en menor grado VAV. Se concluye que la proteína E2 sola y en conjunto con anti-CD81 inducen señales inhibitorias responsables de la disminución en la activación de las células NK de pacientes infectados por el VHC y que éste fenómeno puede ser responsable de la cronicidad que se reporta en dicha enfermedad


The decrease in NK cell activation and cytotoxic activity in patients infected with hepatitis C virus has been described; however, the mechanism by whcih this phenomenon occurs is not known. In the present report, the effect of the E2 protein of the virus envelope or the stimulation of its receptor CD81 with the antibody anti-CD81 on the phosphorylation of tyrosines, serine, the enzymes protein kinase C, phosphoinositol kinase 3 (PI3K), the transcription factor NfkB and the nucleotide exchange protein VAV was assessed in NK cells from normal controls stimulated with anti-CD16. Both the protein E2 and anti-CD81 by themselves or combined, generated a decrease in tyrosine, serine, and a marked decrease in the phosphorylation of PKC, NfkB, PI3k and in less extent in VAV. It is concluded that the E2 protein alone and combined with anti-CD81 induce inhibitory signals responseible for the decrease in the activation of NK cells of infected HCV patients and it could be responsible for the chronicity observed in this disease


Asunto(s)
Humanos , /uso terapéutico , Células Asesinas Naturales/virología , Hepatitis C/terapia , Hepatitis C/virología , Proteína Quinasa C/uso terapéutico , /efectos adversos , /uso terapéutico , Proteínas Proto-Oncogénicas c-vav/uso terapéutico , Receptores de IgG/uso terapéutico , Alergia e Inmunología
12.
Immunology ; 136(1): 46-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22236212

RESUMEN

Systemic lupus erythematosus (SLE) is a multisystem chronic inflammatory disease affecting many organs. The deposition in kidney tissue of immune complexes and their interaction with macrophages is thought to trigger the inflammatory response leading to glomerulonephritis. It has been demonstrated that inhibition of this interaction in murine models can alleviate the disease. Six synthetic peptides were derived from the membrane-proximal extracellular domain (EC2) of human Fcγ receptor II (huFcγRII). Of these, one peptide, huRII6, was shown to be a potent competitive inhibitor of IgG binding to recombinant FcγRII in vitro. To examine the possible therapeutic impact of huRII6 in vivo, this peptide, or a control, was given by subcutaneous injection to female MRL/lpr mice from weeks 7 to 36, resulting in an enhanced survival rate compared with control-treated animals and a reduction of proteinuria. Histopathological examination of the kidneys showed a reduction in deposition of immune complexes and preservation of structure. Such a functional peptide should prove useful for examining the role of IgG-FcγR interactions in experimental models of disease and may provide for the development of FcR-targeting drugs to treat autoimmune disorders.


Asunto(s)
Riñón/lesiones , Fragmentos de Péptidos/inmunología , Receptores de IgG/inmunología , Animales , Células COS , Chlorocebus aethiops , Femenino , Humanos , Inmunoglobulina G/inmunología , Riñón/patología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos MRL lpr , Receptores de IgG/uso terapéutico
13.
J Immunol ; 186(6): 3289-93, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21317388

RESUMEN

Despite their increasing use in autoimmune, inflammatory, and allergic conditions, the mechanism of action of i.v. Igs (IVIg) is poorly understood. On the basis of the critical role of invariant NKT (iNKT) cells in allergic airway inflammation (AAI) and their constitutive expression of the low-affinity IgG receptor FcγRIIIA, we surmised that IVIg targets iNKT cells to exert their anti-inflammatory effect. We found that IVIg treatment significantly inhibited AAI in OVA-sensitized C57BL/6 mice and downregulated α-galactosylceramide-induced iNKT cell activation and cytokine production. Allergic responses were restored in iNKT cell-deficient mice by transferring iNKT cells from PBS- but not from IVIg-treated mice, suggesting that IVIg acts directly on activated iNKT cells that have a critical role in AAI. The inhibitory effects of IVIg on both iNKT cell activation/function and OVA-driven AAI were lost in FcγRIIIA(-/-) mice. Our data unravel an FcγRIIIA-dependent inhibitory effect of IVIg on activated iNKT cells that confers protection in AAI.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Inmunoglobulinas Intravenosas/fisiología , Mediadores de Inflamación/fisiología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/patología , Receptores de IgG/fisiología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/prevención & control , Traslado Adoptivo , Alérgenos/inmunología , Alérgenos/toxicidad , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/terapia , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Galactosilceramidas/antagonistas & inhibidores , Galactosilceramidas/farmacología , Inmunoglobulinas Intravenosas/uso terapéutico , Mediadores de Inflamación/uso terapéutico , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/metabolismo , Ovalbúmina/inmunología , Ovalbúmina/toxicidad , Receptores de IgG/uso terapéutico , Hipersensibilidad Respiratoria/patología , Bazo/inmunología , Bazo/patología , Bazo/trasplante
14.
Allergy ; 66(7): 961-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21272035

RESUMEN

BACKGROUND: Aggregation of the high-affinity IgE receptor (FcεRI) with the low-affinity IgG receptor (FcγRIIb) on basophils or mast cells has been shown to inhibit allergen-induced cell degranulation. Molecules cross-linking these two receptors might therefore be of interest for the treatment of allergic disorders. Here, we demonstrate the generation of a novel bispecific fusion protein efficiently aggregating FcεRI-bound IgE with FcγRIIb on the surface of basophils to prevent pro-inflammatory mediator release. METHODS: Alternative binding molecules recognizing receptor-bound human IgE were selected from DARPin (designed ankyrin repeat protein) libraries. One of the selected DARPins was linked to the Fc-part of a human IgG(1) antibody for binding to FcγRIIb. RESULTS: The resulting anti-IgE DARPin-Fc fusion protein was not anaphylactogenic and inhibited allergen-induced basophil activation in whole blood assays. Both binding moieties of the fusion protein, namely the anti-IgE DARPin as well as the IgG(1) Fc-part, were required to achieve this inhibitory effect. Most importantly, inhibition was faster and more efficient than with Omalizumab, a humanized anti-IgE antibody currently used for the treatment of severe asthma. CONCLUSION: This novel anti-IgE DARPin-Fc fusion protein might represent a potential drug candidate for preventive or immediate treatment of allergic reactions.


Asunto(s)
Hipersensibilidad/inmunología , Proteínas Musculares/uso terapéutico , Proteínas Nucleares/uso terapéutico , Receptores Fc/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Alérgenos/inmunología , Anticuerpos Antiidiotipos/inmunología , Basófilos/inmunología , Degranulación de la Célula/inmunología , Humanos , Hipersensibilidad/tratamiento farmacológico , Inmunoglobulina E/inmunología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Fc/genética , Receptores Fc/inmunología , Receptores Fc/metabolismo , Receptores de IgE/genética , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Receptores de IgE/uso terapéutico , Receptores de IgG/genética , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Receptores de IgG/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
15.
Int J Radiat Oncol Biol Phys ; 78(5): 1513-23, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20932668

RESUMEN

PURPOSE: We determined whether anti-transforming growth factor-ß (TGF-ß) intervention could halt the progression of established radiation-induced liver fibrosis (RILF). METHODS AND MATERIALS: A replication-defective adenoviral vector expressing the extracellular portion of human TßRII and the Fc portion of immunoglobulin G fusion protein (AdTßRIIFc) was produced. The entire rat liver was exposed to 30 Gy irradiation to generate a RILF model (RILFM). Then, RILFM animals were treated with AdTßRIIFc (1 × 10(11) plaque-forming units [PFU] of TßRII), control virus (1 × 10(11) PFU of AdGFP), or saline. Delayed radiation liver injury was assessed by histology and immunohistochemistry. Chronic oxidative stress damage, hepatic stellate cell activation, and hepatocyte regeneration were also analyzed. RESULTS: In rats infected with AdTßRIIFc, fibrosis was significantly improved compared with rats treated with AdGFP or saline, as assessed by histology, hydroxyproline content, and serum level of hyaluronic acid. Compared with AdGFP rats, AdTßRIIFc-treated rats exhibited decreased oxidative stress damage and hepatic stellate cell activation and preserved liver function. CONCLUSIONS: Our results demonstrate that TGF-ß plays a critical role in the progression of liver fibrosis and suggest that anti-TGF-ß intervention is feasible and ameliorates established liver fibrosis. In addition, chronic oxidative stress may be involved in the progression of RILF.


Asunto(s)
Terapia Genética/métodos , Cirrosis Hepática Experimental/prevención & control , Hígado/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adenoviridae/genética , Animales , Vectores Genéticos/uso terapéutico , Células Estrelladas Hepáticas , Ácido Hialurónico/sangre , Hidroxiprolina/análisis , Hígado/metabolismo , Cirrosis Hepática Experimental/metabolismo , Masculino , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/sangre , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/uso terapéutico , Traumatismos Experimentales por Radiación/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de IgG/metabolismo , Receptores de IgG/uso terapéutico , Receptores de Factores de Crecimiento Transformadores beta/sangre , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Factor de Crecimiento Transformador beta/metabolismo , Ensayo de Placa Viral/métodos
16.
Breast ; 18 Suppl 3: S59-63, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19914545

RESUMEN

Treatment decisions for breast cancer patients are currently based on a small number of crude predictive markers, despite the known complexity and heterogeneity of the disease. The field of pharmacogenetics can increase the precision with which therapeutic decisions are made. Discovering associations between genetic variation and treatment response will allow clinicians to tailor therapies to most effectively treat that specific tumor in that patient. In this review we outline two genes with potential clinical relevance in breast cancer treatment. A common polymorphism in the gene encoding Fc fragment of IgG low affinity IIIa receptor (FCGR3A; gene: FCGR3A) may substantially influence a patient's likelihood of responding to trastuzumab. The other gene that will be discussed in the review is cytochrome P450 2D6 (CYP2D6; gene: CYP2D6), which has many genetic variants that impair the bioactivation and effectiveness of tamoxifen therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Citocromo P-450 CYP2D6/genética , Receptores de IgG/genética , Tamoxifeno/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados , Antineoplásicos/metabolismo , Neoplasias de la Mama/genética , Citocromo P-450 CYP2D6/metabolismo , Femenino , Genotipo , Humanos , Farmacogenética , Polimorfismo de Nucleótido Simple , Receptores de IgG/metabolismo , Receptores de IgG/uso terapéutico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Tamoxifeno/metabolismo , Trastuzumab
17.
J Immunol ; 182(11): 7272-9, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454724

RESUMEN

Binding of immune complexes to cellular FcgammaRs can promote cell activation and inflammation. In previous studies, a recombinant human (rh) soluble FcgammaR, rh-FcgammaRIA (CD64A), was shown to block inflammation in passive transfer models of immune complex-mediated disease. To assess whether rh-FcgammaRIA could block inflammation in a T cell- and B cell-dependent model of immune complex-mediated disease, the efficacy of rh-FcgammaRIA in collagen-induced arthritis was evaluated. Mice with established arthritis were treated with a single s.c. injection of rh-FcgammaRIA (0.2-2.0 mg/dose) given every other day for 11 days. Relative to mice injected with vehicle alone, mice treated with rh-FcgammaRIA exhibited lower serum concentrations of IL-6, anti-type II collagen Abs, and total IgG2a. These changes were correlated with lower levels of paw swelling and joint damage in the rh-FcgammaRIA-treated mice and occurred in the presence of a significant murine Ab response to rh-FcgammaRIA. Comparison of the serum rh-FcgammaRIA concentration vs time profiles for rh-FcgammaRIA administered at two dose levels by i.v. and s.c. injection revealed that the bioavailabilty of s.c. administered rh-FcgammaRIA was 27-37%. Taken together, these data show that rh-FcgammaRIA is an effective inhibitor of inflammation in a model of established arthritis in mice.


Asunto(s)
Artritis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Receptores de IgG/administración & dosificación , Animales , Anticuerpos/sangre , Formación de Anticuerpos , Artritis/inducido químicamente , Artritis/patología , Colágeno/efectos adversos , Colágeno/inmunología , Humanos , Inmunoglobulina G/sangre , Interleucina-6/sangre , Ratones , Farmacocinética , Receptores de IgG/uso terapéutico , Proteínas Recombinantes , Solubilidad , Resultado del Tratamiento
18.
Int J Biochem Cell Biol ; 41(3): 472-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18339576

RESUMEN

Transforming growth factor-beta (TGF-beta) is a cytokine that plays a pivotal role in growth, differentiation, development, immune response and wound healing. TGF-beta is upregulated following wound infliction and inflammation, and plays an important role in the production of extracellular matrix proteins that contribute to tissue repair. However, in some diseases, TGF-beta dysregulation can lead to tumor formation, organ fibrosis and the disruption of organ function. A number of molecules have been designed to counteract the effects of TGF-beta, including anti-TGF-beta monoclonal antibodies and various small molecules. Here we discuss the design, use and advantages of the highly specific TGF-beta binding molecule, the soluble human TGF-beta receptor (sTbetaR.Fc) as a TGF-beta sequestering agent.


Asunto(s)
Inmunoterapia/tendencias , Neoplasias/inmunología , Receptores de IgG/inmunología , Proteínas Recombinantes/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos Monoclonales , Diferenciación Celular/inmunología , Proliferación Celular , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Fibrosis , Humanos , Inmunidad Innata , Neoplasias/patología , Neoplasias/terapia , Receptores de IgG/genética , Receptores de IgG/uso terapéutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas/inmunología
19.
Ann Rheum Dis ; 67(2): 154-61, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17557887

RESUMEN

OBJECTIVES: Systemic lupus erythematosus (SLE) is a classical autoimmune disorder characterised by the production of IgG autoantibodies against double-stranded DNA (dsDNA). Activation of Fc gamma R-bearing effector cells by immune complexes (ICs) is a key event in SLE pathogenesis as lupus-prone NZB/NZW F(1) hybrids lacking activating Fc gamma receptors (Fc gamma R) are protected against inflammatory kidney damage despite glomerular deposition of ICs. Moreover, soluble Fc gamma Rs inhibit IC-caused Arthus reaction in vivo. Therefore, recombinant human soluble Fc gamma RII (CD32) was evaluated as a novel therapeutic strategy in lupus-like disease in NZB/NZW F(1) hybrids. METHODS: Binding of husCD32 to murine IgG was studied in vitro by binding to IgG-coated erythrocytes and inhibition of phagocytosis of IgG-opsonised murine erythrocytes. In order to examine therapeutic impact of husCD32 in vivo, female NZB/NZW F(1) mice were treated either from week 16 to 20 ("prophylactic", 150 microg/week husCD32) or continuously from week 24 ("therapeutic"; 100 microg/week husCD32) by subcutaneous injections. Controls received buffered saline. RESULTS: In vitro investigations of husCD32 revealed binding to murine erythrocytes coated with murine IgG. Moreover, husCD32 substantially diminished phagocytosis of murine IgG-opsonised murine red blood cells by peritoneal macrophages indicating disruption of IgG-Fc gamma R interaction. There was a therapeutic efficacy of husCD32 to attenuate lupus pathology indicated by significantly delayed onset of proteinuria and weight loss, reduced histopathological findings, delayed development of anaemia and improved survival by prophylactic application. Therapeutic treatment did not reverse nephritis but significantly prolonged survival despite apparent kidney damage. B cell count, concentration of IgG anti-dsDNA autoantibodies and deposition of glomerular ICs was not significantly affected by the application of husCD32. CONCLUSIONS: The results demonstrate binding properties of husCD32 to ICs in vitro and as a proof-of-principle therapeutic efficacy in inhibiting chronic murine lupus pathology in vivo.


Asunto(s)
Lupus Eritematoso Sistémico/tratamiento farmacológico , Receptores de IgG/uso terapéutico , Animales , Anticuerpos Antinucleares/inmunología , Complejo Antígeno-Anticuerpo/inmunología , ADN/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Inmunoglobulina G/inmunología , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos NZB , Fagocitosis/inmunología , Proteinuria/inmunología , Proteinuria/prevención & control , Receptores de IgG/inmunología , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico
20.
J Immunol ; 180(1): 580-9, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18097060

RESUMEN

Binding of Ag-Ab immune complexes to cellular FcgammaR promotes cell activation, release of inflammatory mediators, and tissue destruction characteristic of autoimmune disease. To evaluate whether a soluble FcgammaR could block the proinflammatory effects of immune complexes, recombinant human (rh) versions of FcgammaRIA, FcgammaRIIA, and FcgammaRIIIA were prepared. Binding of rh-FcgammaRIA to IgG was of high affinity (KD=1.7x10(-10) M), whereas rh-FcgammaRIIA and rh-FcgammaRIIIA bound with low affinity (KD=0.6-1.9x10(-6) M). All rh-FcgammaR reduced immune complex precipitation, blocked complement-mediated lysis of Ab-sensitized RBC, and inhibited immune complex-mediated production of IL-6, IL-13, MCP-1, and TNF-alpha by cultured mast cells. Local or systemic delivery only of rh-FcgammaRIA, however, reduced edema and neutrophil infiltration in the cutaneous Arthus reaction in mice. 125I-labeled rh-FcgammaRIA was cleared from mouse blood with a rapid distribution phase followed by a slow elimination phase with a t1/2gamma of approximately 130 h. The highest percentage of injected radioactivity accumulated in blood approximately liver approximately carcass>kidney. s.c. dosing of rh-FcgammaRIA resulted in lower serum levels of inflammatory cytokines and prevented paw swelling and joint damage in a murine model of collagen Ab-induced arthritis. These data demonstrate that rh-FcgammaRIA is an effective inhibitor of type III hypersensitivity.


Asunto(s)
Enfermedades del Complejo Inmune/tratamiento farmacológico , Receptores de IgG/uso terapéutico , Animales , Complejo Antígeno-Anticuerpo/efectos de los fármacos , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Reacción de Arthus/tratamiento farmacológico , Reacción de Arthus/patología , Proteínas del Sistema Complemento/inmunología , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Humanos , Enfermedades del Complejo Inmune/patología , Inmunoglobulina G/metabolismo , Mastocitos/inmunología , Ratones , Receptores de IgG/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA