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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 109: 37-50, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36581304

RESUMEN

Intravenous immunoglobulin (IVIG) is a promising immune-modulatory therapy for limiting harmful inflammation and associated secondary tissue loss in neurotrauma. Here, we show that IVIG therapy attenuates spatial learning and memory deficits following a controlled cortical impact mouse model of traumatic brain injury (TBI). These improvements in cognitive outcomes were associated with increased neuronal survival, an overall reduction in brain tissue loss, and a greater preservation of neural connectivity. Furthermore, we demonstrate that the presence of the main inhibitory FcγRIIB receptor is required for the beneficial effects of IVIG treatment in TBI, with our results simultaneously highlighting the role of this receptor in reducing secondary damage arising from brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Ratones , Animales , Inmunoglobulinas Intravenosas/farmacología , Inmunoglobulinas Intravenosas/uso terapéutico , Encéfalo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Encefálicas/complicaciones , Cognición
2.
Pharmacology ; 108(2): 176-187, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36696888

RESUMEN

INTRODUCTION: Neutrophils are a pivotal cell type in the K/BxN mouse model of rheumatoid arthritis and play an essential role in the progression of the arthritis. They are readily activated by immune complexes (ICs) via their FcγRs to release IL-1ß in addition to other cytokines, which are inducing cartilage destruction. Neutrophils also release neutrophil-active chemokines to recruit themselves in an autocrine manner to perpetuate tissue destruction. FcγR-expression on neutrophils is of crucial importance for the recognition of ICs. METHODS: In this study, due to its high avidity for binding to FcγRs, we investigated the potential anti-inflammatory effect of a recombinant IgG1 Fc hexamer (rFc-µTP-L309C) on neutrophils in the K/BxN mouse model of endogenously generated chronic arthritis. 200 mg/kg rFc-µTP-L309C and human serum albumin (HSA), used as controls, were administered subcutaneously every other day. Mouse ankle joints were monitored daily to generate a clinical score. Immunohistology was used to evaluate neutrophil infiltration and TUNEL to assess apoptosis. ELISA was used to measure IL-1ß. RESULTS: Treatment with rFc-µTP-L309C, but not HSA, was able to significantly ameliorate the arthritis in the K/BxN mice. Significant neutrophil infiltration into the ankle joint was found, but treatment with rFc-µTP-L309C resulted in significantly less neutrophil infiltration. There was no significant influence of rFc-µTP-L309C on neutrophil death or apoptosis. Less neutrophil infiltration could not be correlated to chemokine-mediated migration. Significantly less IL-1ß was measured in mice treated with rFc-µTP-L309C. CONCLUSION: In the endogenous K/BxN mouse model of rheumatoid arthritis, amelioration can be explained in part by inhibition of neutrophil infiltration into the joints as well as inhibition of IL-1ß production. Given the observed inhibitory properties on neutrophils, rFc-µTP-L309C may be a potential therapeutic candidate to treat autoimmune and inflammatory conditions in which neutrophils are the predominant cell type involved in pathogenesis.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Humanos , Ratones , Animales , Inmunoglobulina G/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patología , Uridina Trifosfato/metabolismo , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Factores Inmunológicos , Ratones Endogámicos C57BL
3.
Eur J Immunol ; 49(1): 195-198, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30267564

RESUMEN

Intravenous immunoglobuin (IVIG) exerts protective effects in experimental allergic bronchopulmonary aspergillosis (ABPA) via a sialylation-dependent mechanism. The protection was associated with reduced recruitment of eosinophils, diminished goblet cell hyperplasia, suppressed Th2 and Th17 responses and reciprocally enhanced regulatory T cells and IL-10, and decreased IgE levels in the circulation.


Asunto(s)
Aspergilosis Broncopulmonar Alérgica/terapia , Eosinófilos/inmunología , Células Caliciformes/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Células Cultivadas , Humanos , Inmunoglobulina E/sangre , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico/metabolismo
4.
J Immunol ; 200(8): 2542-2553, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29531170

RESUMEN

Activation of Fc receptors and complement by immune complexes is a common important pathogenic trigger in many autoimmune diseases and so blockade of these innate immune pathways may be an attractive target for treatment of immune complex-mediated pathomechanisms. High-dose IVIG is used to treat autoimmune and inflammatory diseases, and several studies demonstrate that the therapeutic effects of IVIG can be recapitulated with the Fc portion. Further, recent data indicate that recombinant multimerized Fc molecules exhibit potent anti-inflammatory properties. In this study, we investigated the biochemical and biological properties of an rFc hexamer (termed Fc-µTP-L309C) generated by fusion of the IgM µ-tailpiece to the C terminus of human IgG1 Fc. Fc-µTP-L309C bound FcγRs with high avidity and inhibited FcγR-mediated effector functions (Ab-dependent cell-mediated cytotoxicity, phagocytosis, respiratory burst) in vitro. In addition, Fc-µTP-L309C prevented full activation of the classical complement pathway by blocking C2 cleavage, avoiding generation of inflammatory downstream products (C5a or sC5b-9). In vivo, Fc-µTP-L309C suppressed inflammatory arthritis in mice when given therapeutically at approximately a 10-fold lower dose than IVIG, which was associated with reduced inflammatory cytokine production and complement activation. Likewise, administration of Fc-µTP-L309C restored platelet counts in a mouse model of immune thrombocytopenia. Our data demonstrate a potent anti-inflammatory effect of Fc-µTP-L309C in vitro and in vivo, likely mediated by blockade of FcγRs and its unique inhibition of complement activation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Enfermedades Autoinmunes/inmunología , Proteínas del Sistema Complemento/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Receptores Fc/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular , Activación de Complemento/inmunología , Humanos , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Fagocitosis/inmunología , Receptores de IgG/inmunología
5.
BMC Immunol ; 20(1): 44, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801459

RESUMEN

BACKGROUND: High-dose intravenous immunoglobulin (IVIg), and more recently, subcutaneously-delivered Ig (SCIg), are used to treat a variety of autoimmune diseases; however, there are challenges associated with product production, availability, access and efficacy. These challenges have provided incentives to develop a human recombinant Fc as a more potent alternative to IVIg and SCIg for the treatment of autoimmune diseases. Recently, a recombinant human IgG1 Fc hexamer (Fc-µTP-L309C) was shown to be more efficacious than IVIg in a variety of autoimmune mouse models. We have now examined its efficacy compared to IVIg and SCIg in the K/BxN mouse model of endogenous, chronic rheumatoid arthritis (RA). RESULT: Using the serum-transfer K/BxN model and the endogenous autoimmune model, amelioration of the arthritis was achieved. Effective treatment required high and frequent doses of IVIg, SCIg and Fc-µTP-L309C. However, Fc-µTP-L309C was efficacious at 10-fold lower doses that IVIg/SCIg. Also, arthritis could be prevented when Fc-µTP-L309C was given prior to onset of the arthritis in both the endogenous model and in the serum transfer model. CONCLUSIONS: Our results show that Fc-µTP-L309C is a powerful treatment for the prevention and amelioration of severe, chronic arthritis in a true autoimmune mouse model of RA. Thus, the K/BxN endogenous arthritis model should be useful for testing potential therapeutics for RA. Our findings provide rationale for further examination of the treatment efficacy of immunoglobulin-based therapeutics in rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Animales , Artritis Reumatoide/patología , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Inmunoglobulina G/química , Inmunoglobulinas Intravenosas/administración & dosificación , Factores Inmunológicos/administración & dosificación , Ratones , Resultado del Tratamiento
6.
Respir Res ; 20(1): 99, 2019 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118031

RESUMEN

BACKGROUND: Recurrent and persistent infections are known to affect airways of patients with Primary Immunodeficiency despite appropriate replacement immunoglobulin serum levels. Interestingly, patients with Chronic Obstructive Pulmonary Disease or with non-CF bronchiectasis also show similar susceptibility to such infections. This may be due to the limited availability of immunoglobulins from the systemic circulation in the conductive airways, resulting in local immunodeficiency. Topical application of nebulized plasma-derived immunoglobulins may represent a means to address this deficiency. In this study, we assessed the feasibility of nebulizing plasma-derived immunoglobulins and delivering them into the airways of rats and non-human primates. METHODS: Distinct human plasma-derived immunoglobulin isotype preparations were nebulized with an investigational eFlow® nebulizer and analyzed in vitro or deposited into animals. Biochemical and immunohistological analysis of nebulized immunoglobulins were then performed. Lastly, efficacy of topically applied human plasma-derived immunoglobulins was assessed in an acute Streptococcus pneumoniae respiratory infection in mice. RESULTS: Characteristics of the resulting aerosols were comparable between preparations, even when using solutions with elevated viscosity. Neither the structural integrity nor the biological function of nebulized immunoglobulins were compromised by the nebulization process. In animal studies, immunoglobulins levels were assessed in plasma, broncho-alveolar lavages (BAL) and on lung sections of rats and non-human primates in samples collected up to 72 h following application. Nebulized immunoglobulins were detectable over 48 h in the BAL samples and up to 72 h on lung sections. Immunoglobulins recovered from BAL fluid up to 24 h after inhalation remained structurally and functionally intact. Importantly, topical application of human plasma-derived immunoglobulin G into the airways of mice offered significant protection against acute pneumococcal pneumonia. CONCLUSION: Taken together our data demonstrate the feasibility of topically applying plasma-derived immunoglobulins into the lungs using a nebulized liquid formulation. Moreover, topically administered human plasma-derived immunoglobulins prevented acute respiratory infection.


Asunto(s)
Inmunoglobulina A/administración & dosificación , Inmunoglobulina G/administración & dosificación , Inmunoglobulina M/administración & dosificación , Pulmón/efectos de los fármacos , Nebulizadores y Vaporizadores/tendencias , Administración Tópica , Animales , Relación Dosis-Respuesta a Droga , Humanos , Inmunoglobulina A/metabolismo , Inmunoglobulina G/metabolismo , Pulmón/metabolismo , Macaca fascicularis , Ratones Endogámicos C57BL , Ratones Transgénicos , Primates , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
7.
J Immunol ; 192(11): 5031-8, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24760152

RESUMEN

High-dose i.v. Ig (IVIG) is used to treat various autoimmune and inflammatory diseases; however, the mechanism of action remains unclear. Based on the K/BxN serum transfer arthritis model in mice, IVIG suppression of inflammation has been attributed to a mechanism involving basophils and the binding of highly sialylated IgG Fc to DC-SIGN-expressing myeloid cells. The requirement for sialylation was examined in the collagen Ab-induced arthritis (CAbIA) and K/BxN serum transfer arthritis models in mice. High-dose IVIG (1-2 g/kg body weight) suppressed inflammatory arthritis when given prophylactically. The same doses were also effective in the CAbIA model when given subsequent to disease induction. In this therapeutic CAbIA model, the anti-inflammatory effect of IVIG was dependent on IgG Fc but not F(ab')2 fragments. Removal of sialic acid residues by neuraminidase had no impact on the anti-inflammatory activity of IVIG or Fc fragments. Treatment of mice with basophil-depleting mAbs did not abrogate the suppression of either CAbIA or K/BxN arthritis by IVIG. Our data confirm the therapeutic benefit of IVIG and IgG Fc in Ab-induced arthritis but fail to support the significance of sialylation and basophil involvement in the mechanism of action of IVIG therapy.


Asunto(s)
Artritis/inmunología , Artritis/prevención & control , Basófilos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Ácido N-Acetilneuramínico/inmunología , Animales , Artritis/patología , Basófilos/patología , Modelos Animales de Enfermedad , Inmunoglobulinas Intravenosas/inmunología , Factores Inmunológicos/inmunología , Masculino , Ratones , Ratones Endogámicos NOD
8.
Clin Immunol ; 161(2): 373-83, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26307433

RESUMEN

Intravenous IgG (IVIG) therapy can be used for immunomodulation. IL-2 is an immunoregulatory cytokine. We evaluated IVIG modulation of human blood lymphocyte response to IL-2 and other cytokines. Neither IVIG nor low concentrations of IL-2 (3-30U/ml) induced lymphocyte proliferation, but in combination they synergistically enhanced proliferation of NK cells. The CD56(bright) cells expanded more than CD56(dim) NK cells, with 90% of NK cells dividing up to 8 generations by day 6, while <8% of T cells divided. IVIG also potentiated NK cell proliferation with IL-12, IL-15 and IL-18. The IVIG+cytokine-expanded NK cells were less cytotoxic for K562 cells, than NK cells with cytokine alone. IVIG also enhanced interferon-γ production with IL-2, IL-12 and IL-18. In conclusion, IVIG selectively potentiates NK cell proliferation and interferon-γ secretion with IL-2, IL-12, IL-15 and IL-18 in vitro. These findings warrant evaluation in vivo in relation to NK cells and the immunoregulatory actions of IVIG.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Citocinas/farmacología , Inmunoglobulinas Intravenosas/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Factores Inmunológicos/farmacología , Interferón gamma/metabolismo , Interleucina-12/farmacología , Interleucina-15/farmacología , Interleucina-18/farmacología , Interleucina-2/farmacología , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo
9.
Clin Immunol ; 160(2): 123-32, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25982320

RESUMEN

IVIG modulates T cell activation in vitro and inflammatory-autoimmune conditions in vivo. Sialylation of IgG, Fc receptor interactions, modulation of monocyte/macrophage/B cell functions have been implicated in IVIG effects. Subcutaneous IgG (SCIG) therapy is increasingly used for IgG replacement but whether these preparations share the effects of IVIG on T cell modulation is not documented. We compared the potency of SCIG-Hizentra™ (20% IgG preparation) with IVIG-Privigen® (10% IgG) for T cell inhibition, and assessed the involvement of IgG sialylation, monocytes and B cells in this process. Human PBMCs or sorted cells were cultured 3-7 days, and T cells were stimulated with immobilized anti-CD3 mAb or Candida antigen. Thymidine incorporation into DNA was quantitated and cytokines assayed by ELISA/Luminex® assay. IVIG and SCIG both dose-dependently (1-20mg/ml) inhibited (up to >80%) T cell proliferation to anti-CD3 mAb. Response to Candida albicans was comparably inhibited by IVIG and SCIG by 50-80% at 10mg/ml with inhibition even at 3mg/ml (P<0.05). These effects were not affected by depletion of sialic acid containing IgG using neuraminidase treatment or lectin affinity chromatography. With anti-CD3 or Candida stimulation, IL-1ß, IL-2, IL-5, IL-6, IL-13, GMCSF, TNF-α, interferon-γ (with anti-CD3) and IL-17 (with Candida) levels were suppressed by IVIG or SCIG, with no effect on IL-4, IL-10, IL-12, IL-15 or TGFß. Monocytes or B cells were not required for IgG-induced suppression of proliferation, in fact depletion of monocytes potentiated the IgG-induced inhibition. Reconstitution with monocytes restored the original inhibitory effect. These data show that IVIG (Privigen®) and SCIG (Hizentra™) have comparable inhibitory effects on T cell activation, which do not require sialylation of IgG. Inhibition is independent of monocytes or B cells. There is a potent suppression of multiple effector cytokines. Like IVIG, SCIG therapy is expected to show immunomodulatory activity.


Asunto(s)
Linfocitos B/efectos de los fármacos , Citocinas/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Antígenos Fúngicos/inmunología , Linfocitos B/inmunología , Complejo CD3/inmunología , Candida albicans/inmunología , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/farmacología , Inmunoglobulinas Intravenosas/administración & dosificación , Factores Inmunológicos/administración & dosificación , Técnicas In Vitro , Infusiones Subcutáneas , Activación de Linfocitos/inmunología , Ácido N-Acetilneuramínico , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Eur J Immunol ; 44(7): 2059-63, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700174

RESUMEN

Several mechanisms account for the beneficial effect of intravenous immunoglobulin (IVIg) in autoimmune and inflammatory diseases. These mechanisms include effects on the cellular compartment and on the humoral compartment. Thus, IVIg impacts on dendritic cells, macrophages, neutrophils, basophils, NK cells, and B and T lymphocytes. Several studies have emphasized that the antiinflammatory effect of IVIg is dependent on α2,6-sialylation of the N-linked glycan on asparagine-297 of the Fc portion of IgG. However, recent reports have questioned the necessity of sialylated Fc and the role of FcγRIIB in IVIg-mediated antiinflammatory effects. In view of the critical role played by Th17 cells in several autoimmune pathologies and the increasing use of IVIg in several of these conditions, by using neuraminidase-treated, desialylated IVIg, we addressed whether the α2,6-sialylation of IgG is essential for the beneficial effect of IVIg in experimental autoimmune encephalomyelitis (EAE), a Th17-driven condition, and for the reciprocal modulation of helper T-cell subsets. We observed no difference in the ability of IVIg to ameliorate EAE irrespective of its sialylation. Our findings thus show that sialylation of IVIg is not necessary for IVIg-mediated amelioration of EAE or for downregulation of Th17 cells and upregulation of regulatory T cells.


Asunto(s)
Inmunoglobulinas Intravenosas/farmacología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/etiología , Femenino , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulinas Intravenosas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Ácido N-Acetilneuramínico/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células Th17/fisiología
11.
Arthritis Rheum ; 65(10): 2713-23, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23840006

RESUMEN

OBJECTIVE: Plasmacytoid dendritic cells (PDCs) produce high concentrations of interferon-α (IFNα) following exposure to immune complexes containing nucleic acids. We previously reported that serum from healthy donors inhibits IFNα production by PDCs in response to systemic lupus erythematosus (SLE) immune complexes, and that inhibition is mediated, in part, by IgG. IgG is the major component of intravenous immunoglobulin and is well known to exert antiinflammatory properties. Although suppression of inflammation by the sialylated subfraction of IgG has been implicated in some models, the mechanism of IFNα inhibition by IgG and the importance of sialylation have not been studied. METHODS: SLE immune complexes or synthetic Toll-like receptor (TLR) agonists were used to stimulate total or individual cell-depleted human mononuclear cell cultures in the presence or absence of IgG, Fc fragments, F(ab')2 fragments, and their sialylated or unsialylated subfractions. Cytokines were quantified by enzyme-linked immunosorbent assay. RESULTS: We identified 2 distinct mechanisms by which IgG inhibits IFNα production. First, IgG Fc fragments inhibited SLE immune complex-stimulated IFNα production via a sialic acid-independent mechanism, by inhibiting immune complex binding to Fcγ receptor IIa on PDCs. In contrast, the F(ab')2 fragment of the sialylation-enriched fraction of IgG inhibited TLR-7 or TLR-9 agonist-induced IFNα production but did not require the sialic acid residue itself. The inhibitory activity of IgG on TLR agonist-induced IFNα required monocyte production of prostaglandin E2, a potent suppressor of IFNα production by PDCs. CONCLUSION: IgG attenuates IFNα production by PDCs by both cell surface receptor and intracellular pathways, depending on the nature of the inducing stimulus.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Células Dendríticas/metabolismo , Inmunoglobulina G/farmacología , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/efectos de los fármacos , Receptores Toll-Like/metabolismo , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Dinoprostona/metabolismo , Humanos , Inmunoglobulina G/administración & dosificación , Inyecciones Intravenosas , Interferón-alfa/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Receptores de IgG/metabolismo , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/agonistas
12.
Transfusion ; 52(8): 1799-805, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22257295

RESUMEN

BACKGROUND: Sialylation of the N-linked glycan on asparagine-297 within the Fc region of intravenous gammaglobulins (IVIgs) was shown to be necessary for efficacy of IVIg in the amelioration of experimental inflammatory arthritis. To test the role for Fc sialylation of IVIg in immune modulating therapies beyond the K/BxN arthritis model, we examined the efficacy of sialylated compared to nonsialylated IVIg for the ability to attenuate immune thrombocytopenia (ITP) in a mouse model that approximates the clinical setting of human ITP. STUDY DESIGN AND METHODS: We used a published, passive anti-platelet (PLT) dose-escalation mouse model of ITP that approximates clinical ITP. PLT counts were followed over time before and after IVIg treatment. IVIg from two different manufacturers was used to prepare untreated and neuraminidase-treated IVIg. Sambucus nigra agglutinin (SNA) affinity chromatography was used to obtain sialic acid-enriched and -depleted IVIg. Sialic acid content was determined using Western blot, enzyme-linked immunosorbent assay, and high-performance liquid chromatography. RESULTS: Results were the same using sialylated and desialylated (neuraminidase-treated) IVIg from two different manufacturers. No differences were observed between sialic acid-enriched and -depleted IVIg compared to normal IVIg in their efficacy to alleviate ITP. Using quantitative reverse transcription-polymerase chain reaction, no increase in the spleen FcγRIIB mRNA was detectable, but a pronounced increase of FcγRIIB mRNA in the marrow was seen after IVIg administration. CONCLUSIONS: We conclude that IVIg ameliorates experimental ITP by a mechanism that is independent of sialylation either in the Fc or the Fab region of IVIg.


Asunto(s)
Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Ácido N-Acetilneuramínico/metabolismo , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Púrpura Trombocitopénica Idiopática/inmunología , Animales , Asparagina/metabolismo , Modelos Animales de Enfermedad , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Lectinas de Plantas/farmacología , Recuento de Plaquetas , Polisacáridos/metabolismo , Púrpura Trombocitopénica Idiopática/metabolismo , ARN Mensajero/metabolismo , Receptores de IgG/genética , Proteínas Inactivadoras de Ribosomas/farmacología
13.
EJHaem ; 2(4): 789-793, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35845218

RESUMEN

Recombinant Fc-µTP-L309C is more efficacious than intravenous immunoglobulin (IVIg) at ameliorating antibody-mediated autoimmune diseases through its effects on Fcγ receptors (FcγRs). Fc-µTP-L309C inhibited in-vitro FcγR-mediated phagocytosis 104/105-fold better than IVIg. Fc-µTP-L309C, given subcutaneously, recovered platelet counts in an immune thrombocytopenia (ITP) mouse model to a higher degree than IVIg at a 10-fold lower dose. We show, using confocal microscopy, that Fc-µTP-L309C binds to monocyte-macrophages and is rapidly internalized, whereas, IVIg remains on the cell surface. Western blotting showed that internalized FcγRIII is degraded through a lysosomal pathway, and this reduction of cell surface FcγRIII is likely responsible for the increased efficacy to ameliorate ITP.

14.
Front Immunol ; 12: 660506, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093549

RESUMEN

Intravenous immunoglobulin (IVIG) is an effective immunomodulatory treatment for immune dysregulation diseases. However, the mechanisms by which it reduces systemic inflammation are not well understood. NK cell cytotoxicity is decreased by IVIG in women with reduced fertility, but IVIG effects on NK cells in immune dysregulation are less clear. We hypothesized that IVIG modulation of lymphocyte function, especially in NK cells, is important for resolution of inflammation. Our aim was to identify IVIG-induced changes in a cohort of patients with Kawasaki disease (KD) and those that occur broadly in pediatric patients with various immune dysregulatory diseases. Peripheral blood mononuclear cells (PBMCs) of patients with KD or autoimmune/inflammatory diseases were phenotyped pre and post high dose IVIG treatment by flow cytometry. In KD patients, after IVIG infusion Treg cell frequency and the proportion of activated CD25+ immunoregulatory CD56bright NK cells was increased, and multiple lymphocyte subsets showed increased expression of the lymphoid tissue homing receptor CD62L. Importantly, IVIG treatment decreased the frequency of cells expressing the degranulation marker CD107a among cytotoxic CD56dim NK cells, which was reflected in a significant reduction in target cell killing and in decreased production of multiple pro-inflammatory mediators. Interestingly, the activating receptor CD336 was expressed on a higher proportion of CD56bright NK cells after IVIG in both KD and autoimmune/inflammatory patients while other NK receptors were increased differentially in each cohort. In autoimmune/inflammatory patients IVIG induced the proliferation marker CD71 on a higher percentage of CD56dim NK cells, and in contrast to KD patients, CD107a+ cells were increased in this subset. Furthermore, when PBMCs were stimulated ex vivo with IL-2 or Candida antigen in autologous plasma, more of the CD4+ T cells of KD patients expressed CD25 after IVIG therapy but fewer cytotoxic T cells were degranulated based on CD107a expression. In summary, IVIG treatment in patients with immune dysregulation has multiple effects, especially on NK cell subsets and CD4+ T cells, which are compatible with promoting resolution of inflammation. These novel findings provide insight into the immunomodulatory actions of IVIG in autoimmune and inflammatory conditions.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Inmunoglobulinas Intravenosas/administración & dosificación , Inmunoglobulinas Intravenosas/uso terapéutico , Inflamación/terapia , Células Asesinas Naturales/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Adolescente , Linfocitos T CD4-Positivos/inmunología , Niño , Preescolar , Estudios de Cohortes , Femenino , Citometría de Flujo , Humanos , Inmunomodulación , Células Asesinas Naturales/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Linfocitos T Reguladores/inmunología
15.
Commun Biol ; 3(1): 96, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132640

RESUMEN

Therapeutic normal IgG intravenous immunoglobulin (IVIG) is a well-established first-line immunotherapy for many autoimmune and inflammatory diseases. Though several mechanisms have been proposed for the anti-inflammatory actions of IVIG, associated signaling pathways are not well studied. As ß-catenin, the central component of the canonical Wnt pathway, plays an important role in imparting tolerogenic properties to dendritic cells (DCs) and in reducing inflammation, we explored whether IVIG induces the ß-catenin pathway to exert anti-inflammatory effects. We show that IVIG in an IgG-sialylation independent manner activates ß-catenin in human DCs along with upregulation of Wnt5a secretion. Mechanistically, ß-catenin activation by IVIG requires intact IgG and LRP5/6 co-receptors, but FcγRIIA and Syk are not implicated. Despite induction of ß-catenin, this pathway is dispensable for anti-inflammatory actions of IVIG in vitro and for mediating the protection against experimental autoimmune encephalomyelitis in vivo in mice, and reciprocal regulation of effector Th17/Th1 and regulatory T cells.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , beta Catenina/metabolismo , Animales , Antiinflamatorios/farmacología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/efectos de los fármacos
16.
Cell Death Dis ; 11(1): 50, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974400

RESUMEN

Autophagy plays an important role in the regulation of autoimmune and autoinflammatory responses of the immune cells. Defective autophagy process is associated with various autoimmune and inflammatory diseases. Moreover, in many of these diseases, the therapeutic use of normal immunoglobulin G or intravenous immunoglobulin (IVIG), a pooled normal IgG preparation, is well documented. Therefore, we explored if IVIG immunotherapy exerts therapeutic benefits via induction of autophagy in the immune cells. Here we show that IVIG induces autophagy in peripheral blood mononuclear cells (PBMCs). Further dissection of this process revealed that IVIG-induced autophagy is restricted to inflammatory cells like monocytes, dendritic cells, and M1 macrophages but not in cells associated with Th2 immune response like M2 macrophages. IVIG induces autophagy by activating AMP-dependent protein kinase, beclin-1, class III phosphoinositide 3-kinase and p38 mitogen-activated protein kinase and by inhibiting mammalian target of rapamycin. Mechanistically, IVIG-induced autophagy is F(ab')2-dependent but sialylation independent, and requires endocytosis of IgG by innate cells. Inhibition of autophagy compromised the ability of IVIG to suppress the inflammatory cytokines in innate immune cells. Moreover, IVIG therapy in inflammatory myopathies such as dermatomyositis, antisynthetase syndrome and immune-mediated necrotizing myopathy induced autophagy in PBMCs and reduced inflammatory cytokines in the circulation, thus validating the translational importance of these results. Our data provide insight on how circulating normal immunoglobulins maintain immune homeostasis and explain in part the mechanism by which IVIG therapy benefits patients with autoimmune and inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Autofagia/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , Leucocitos Mononucleares/metabolismo , Adenilato Quinasa/metabolismo , Beclina-1/metabolismo , Línea Celular , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Endocitosis/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Orgánulos/efectos de los fármacos , Orgánulos/metabolismo , Orgánulos/ultraestructura , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Donantes de Tejidos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Autoimmun Rev ; 18(10): 102366, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31404703

RESUMEN

In recent years, there has been a surge in the research and development of novel molecules as potential therapeutic alternatives to traditional treatments (such as intravenous immunoglobulins) for autoimmune disorders. The aim of this review is to describe different drug development strategies and evaluate how various molecules have performed in clinical trials to date. Broadly, three main approaches have been pursued. Recombinant fragment crystallisable (rFc) multimers primarily target Fcγ receptors (FcγRs) but may also affect the complement system. These include PF-06755347 (GL-2045), CSL730 (M230), CSL777 and Pan Fc Receptor Interacting Molecule (PRIM). Neonatal Fc receptor (FcRn)-targeting therapeutics block the FcRn receptor and are represented by candidate drugs such as the Fc fragment efgartigimod and the monoclonal antibodies rozanolixizumab (UCB7665), M281 and SYNT001. Finally, Fc and FcγR-targeting therapeutics, comprise molecules that target the Fc of IgG, such as the recombinant soluble FcγIIb receptor valziflocept (SM101/SHP652) and various monoclonal antibodies directed against the receptors. The developmental status of these three classes of molecules ranges from preclinical to ongoing phase 3 clinical studies. Efgartigimod and rozanolixizumab are the most advanced and have demonstrated encouraging results from phase 2 trials in immune thrombocytopenia and myasthenia gravis. Although initial results are promising, further long-term data and a better understanding of the unique mechanisms of action of the different molecules are needed. The efficacy, safety, convenience of administration, duration of effects, and cost will all contribute to determining which of the molecules will be successful in the clinic.


Asunto(s)
Enfermedades Autoinmunes/terapia , Productos Biológicos/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/inmunología , Terapia Molecular Dirigida , Receptores Fc/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Receptores Fc/inmunología
18.
Mucosal Immunol ; 12(4): 1013-1024, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31105268

RESUMEN

Recurrent and persistent airway infections remain prevalent in patients with primary immunodeficiency (PID), despite restoration of serum immunoglobulin levels by intravenous or subcutaneous plasma-derived IgG. We investigated the effectiveness of different human Ig isotype preparations to protect mice against influenza when delivered directly to the respiratory mucosa. Four polyvalent Ig preparations from pooled plasma were compared: IgG, monomeric IgA (mIgA), polymeric IgA-containing IgM (IgAM) and IgAM associated with the secretory component (SIgAM). To evaluate these preparations, a transgenic mouse expressing human FcαRI/CD89 within the myeloid lineage was created. CD89 was expressed on all myeloid cells in the lung and blood except eosinophils, reflecting human CD89 expression. Intranasal administration of IgA-containing preparations was less effective than IgG in reducing pulmonary viral titres after infection of mice with A/California/7/09 (Cal7) or the antigenically distant A/Puerto Rico/8/34 (PR8) viruses. However, IgA reduced weight loss and inflammatory mediator expression. Both IgG and IgA protected mice from a lethal dose of PR8 virus and for mIgA, this effect was partially CD89 dependent. Our data support the beneficial effect of topically applied Ig purified from pooled human plasma for controlling circulating and non-circulating influenza virus infections. This may be important for reducing morbidity in PID patients.


Asunto(s)
Antígenos CD/genética , Expresión Génica , Isotipos de Inmunoglobulinas/inmunología , Receptores Fc/genética , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos CD/inmunología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Isotipos de Inmunoglobulinas/administración & dosificación , Inmunofenotipificación , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Pruebas de Neutralización , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Unión Proteica/inmunología , Receptores Fc/inmunología
19.
Sci Transl Med ; 11(506)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434758

RESUMEN

Treatment of autoimmune and inflammatory diseases typically involves immune suppression. In an opposite strategy, we show that administration of the highly inflammatory erythrocyte-specific antibody Ter119 into mice remodels the monocyte cellular landscape, leading to resolution of inflammatory disease. Ter119 with intact Fc function was unexpectedly therapeutic in the K/BxN serum transfer model of arthritis. Similarly, it rapidly reversed clinical disease progression in collagen antibody-induced arthritis (CAIA) and collagen-induced arthritis and completely corrected CAIA-induced increase in monocyte Fcγ receptor II/III expression. Ter119 dose-dependently induced plasma chemokines CCL2, CCL5, CXCL9, CXCL10, and CCL11 with corresponding alterations in monocyte percentages in the blood and liver within 24 hours. Ter119 attenuated chemokine production from the synovial fluid and prevented the accumulation of inflammatory cells and complement components in the synovium. Ter119 could also accelerate the resolution of hypothermia and pulmonary edema in an acute lung injury model. We conclude that this inflammatory anti-erythrocyte antibody simultaneously triggers a highly efficient anti-inflammatory effect with broad therapeutic potential.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Eritrocitos/inmunología , Inflamación/tratamiento farmacológico , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/complicaciones , Anemia/sangre , Anemia/complicaciones , Animales , Artritis/sangre , Artritis/complicaciones , Artritis Experimental/sangre , Artritis Experimental/complicaciones , Artritis Experimental/inmunología , Transfusión Sanguínea , Movimiento Celular , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glicosilación , Inmunoglobulina G/metabolismo , Inflamación/sangre , Inflamación/complicaciones , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones SCID , Monocitos/metabolismo , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/complicaciones , Púrpura Trombocitopénica Idiopática/patología , Receptores de IgG/metabolismo
20.
Sci Rep ; 7(1): 1296, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28465620

RESUMEN

Intravenous immunoglobulin (IVIG) are purified IgG preparations made from the pooled plasma from thousands of healthy donors and are being tested in preclinical mouse models. Inherent challenges, however, are the pluripotency of IVIG and its xenogeneicity in animals. IVIG can alter the viability of human neutrophils via agonistic antibodies to Fas and Siglec-9. In this study, we compared the effects of IVIG on human and mouse neutrophils using different death assays. Different commercial IVIG preparations similarly induced cytokine-dependent death in human neutrophils, whereas they had no effects on the survival of either peripheral blood or bone marrow neutrophils from C57BL/6 or BALB/c mice. F(ab')2 but not Fc fragments of IVIG induced death of human neutrophils, whereas neither of these IVIG fragments, nor agonistic monoclonal antibodies to human Fas or Siglec-9 affected the viability of mouse neutrophils. Pooled mouse IgG, which exhibited a different immunoprofile compared to IVIG, also had no effect on mouse cells. Together, these observations demonstrate that effects of IVIG on neutrophil survival are not adequately reflected in current mouse models, despite the key role of these cells in human inflammatory and autoimmune diseases.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Inmunoglobulina G/inmunología , Inmunoglobulinas Intravenosas/administración & dosificación , Neutrófilos/efectos de los fármacos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos CD/inmunología , Supervivencia Celular/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/administración & dosificación , Inmunoglobulinas Intravenosas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/inmunología , Receptor fas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA