Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Arthritis Res Ther ; 22(1): 264, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33168076

RESUMEN

BACKGROUND: Both humoral and cellular immune mechanisms are involved in the onset and progression of autoimmune responses in systemic lupus erythematosus (SLE). Plasmacytoid dendritic cells (pDCs) play a central role in the pathogenesis of SLE via the dysregulation of type I interferon (IFN) production; these cells act together with activated myeloid DCs (mDCs) to amplify the vicious pathogenic spiral of autoimmune disorders. Therefore, control of aberrant DC activation in SLE may provide an alternative treatment strategy against this disease. Mycophenolate mofetil (MMF), which has been used to treat lupus nephritis, specifically blocks the proliferation of B and T lymphocytes via inhibition of inosine-5-monophosphate dehydrogenase. Here, we focus on the effects of MMF in targeting DC functions, especially the IFN response of pDCs. METHODS: We isolated human blood pDCs and mDCs by flow cytometry and examined the effect of mycophenolic acid (MPA), which is a metabolic product of MMF, on the toll-like receptor (TLR) ligand response of DC subsets. Additionally, we cultured pDCs with serum from SLE patients in the presence or absence of MPA and then examined the inhibitory function of MPA on SLE serum-induced IFN-α production. RESULTS: We found that treatment with 1-10 µM of MPA (covering the clinical trough plasma concentration range) dose-dependently downregulated the expression of CD80 and CD86 on mDCs (but not pDCs) without inducing apoptosis, in response to R848 or CpG-ODN, respectively. Notably, in pDCs, MPA significantly suppressed IFN-α production with IRF7 nuclear translocation and repressed the AKT activity. In addition, MPA inhibited IL-12 production with STAT4 expression in mDCs. We further identified that MPA had an inhibitory effect on SLE serum-induced IFN-α production by pDCs. CONCLUSIONS: Our data suggest that MPA can interrupt the vicious pathogenic spiral of autoimmune disorders by regulating the function of DC subsets. This work unveiled a novel mechanism for the therapeutic ability of MMF against SLE.


Asunto(s)
Interferón-alfa , Lupus Eritematoso Sistémico , Ácido Micofenólico , Células Dendríticas , Humanos , Interferón-alfa/efectos de los fármacos , Interferón-alfa/metabolismo , Ácido Micofenólico/farmacología , Linfocitos T
2.
Diagnosis (Berl) ; 7(4): 385-386, 2020 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-32549125

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2), shares similarities with the former SARS outbreak, which was caused by SARS-CoV-1. SARS was characterized by severe lung injury due to virus-induced cytopathic effects and dysregulated hyperinflammatory state. COVID-19 has a higher mortality rate in men both inside and outside China. In this opinion paper, we describe how sex-specific immunobiological factors and differences in angiotensin converting enzyme 2 (ACE2) expression may explain the increased severity and mortality of COVID-19 in males. We highlight that immunomodulatory treatment must be tailored to the underlying immunobiology at different stages of disease. Moreover, by investigating sex-based immunobiological differences, we may enhance our understanding of COVID-19 pathophysiology and facilitate improved immunomodulatory strategies.


Asunto(s)
Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Enzima Convertidora de Angiotensina 2 , Animales , Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Recuento de Linfocito CD4/estadística & datos numéricos , COVID-19 , Infecciones por Coronavirus/terapia , Infecciones por Coronavirus/virología , Citocinas/metabolismo , Femenino , Humanos , Inmunomodulación , Interferón-alfa/efectos de los fármacos , Interferón-alfa/inmunología , Linfopenia/mortalidad , Masculino , Glicoproteínas de Membrana/genética , Ratones , Modelos Animales , Pandemias , Neumonía Viral/terapia , Neumonía Viral/virología , SARS-CoV-2 , Factores Sexuales , Receptor Toll-Like 7/genética
4.
Am J Respir Crit Care Med ; 201(5): 526-539, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31710506

RESUMEN

Rationale: IL-18 is a member of the IL-1 cytokine family, and elevated blood IL-18 concentrations associate with disease activity in macrophage activation syndrome (MAS) and poor clinical outcomes in severe inflammatory and septic conditions.Objectives: Although recent investigations provide mechanistic evidence for a contribution of IL-18 to inflammation and hyperinflammation in sepsis and MAS, we sought to study regulatory mechanisms underlying human IL-18 expression.Methods: Samples from in vivo and in vitro endotoxin rechallenge experiments, patients with inflammatory disease, and isolated human monocytes treated with various stimulants and drugs were tested for cytokine gene and protein expression. Serum IL-18 expression with or without JAK/STAT inhibition was analyzed in two MAS mouse models and in a patient with recurrent MAS.Measurements and Main Results: Peripheral blood and monocytic IL-18 expression escaped LPS-induced immunoparalysis. LPS-stimulated primary human monocytes revealed specific IL-18 expression kinetics controlled by IFNα/ß signaling. JAK/STAT inhibition or IFNß neutralization during LPS stimulation blunted cytokine expression. Similarly, microtubule-destabilizing drugs abrogated LPS-induced IL18 expression, but this effect could be fully reversed by addition of IFNα/ß. Ex vivo analysis of inflammatory disease patients' whole blood revealed strong correlation of type I IFN score and IL18 expression, whereas JAK/STAT inhibition strongly reduced IL-18 serum levels in two MAS mouse models and in a patient with recurrent MAS.Conclusions: Our data indicate that IL-18 (but not IL-1ß) production from human monocytes requires cooperative Toll-like receptor and IFNα/ß signaling. Interference with IFNα/ß expression or signaling following JAK/STAT inhibition may control catastrophic hyperinflammation in MAS.


Asunto(s)
Tolerancia Inmunológica/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Interleucina-18/inmunología , Síndrome de Activación Macrofágica/inmunología , Receptores Toll-Like/inmunología , Adulto , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Modelos Animales de Enfermedad , Endotoxinas , Expresión Génica , Humanos , Técnicas In Vitro , Interferón-alfa/efectos de los fármacos , Interferón beta/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Inhibidores de las Cinasas Janus/farmacología , Lipopolisacáridos/farmacología , Síndrome de Activación Macrofágica/genética , Síndrome de Activación Macrofágica/metabolismo , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Piperidinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Transducción de Señal , Inhibidores del Factor de Necrosis Tumoral/farmacología
5.
PLoS One ; 14(4): e0215062, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30990863

RESUMEN

Interferon α (IFNα) counteracts viral infections by activating various IFNα-stimulated genes (ISGs). These genes encode proteins that block viral transport into the host cell and inhibit viral replication, gene transcription and translation. Due to the existence of 14 different, highly homologous isoforms of mouse IFNα, an IFNα knockout mouse has not yet been established by genetic knockout strategies. An scFv intrabody for holding back IFNα isoforms in the endoplasmic reticulum (ER) and thus counteracting IFNα secretion is reported. The intrabody was constructed from the variable domains of the anti-mouse IFNα rat monoclonal antibody 4EA1 recognizing the 5 isoforms IFNα1, IFNα2, IFNα4, IFNα5, IFNα6. A soluble form of the intrabody had a KD of 39 nM to IFNα4. It could be demonstrated that the anti-IFNα intrabody inhibits clearly recombinant IFNα4 secretion by HEK293T cells. In addition, the secretion of IFNα4 was effectively inhibited in stably transfected intrabody expressing RAW 264.7 macrophages and dendritic D1 cells. Colocalization of the intrabody with IFNα4 and the ER marker calnexin in HEK293T cells indicated complex formation of intrabody and IFNα4 inside the ER. Intracellular binding of intrabody and antigen was confirmed by co-immunoprecipitation. Complexes of endogenous IFNα and intrabody could be visualized in the ER of Poly (I:C) stimulated RAW 264.7 macrophages and D1 dendritic cells. Infection of macrophages and dendritic cells with the vesicular stomatitis virus VSV-AV2 is attenuated by IFNα and IFNß. The intrabody increased virus proliferation in RAW 264.7 macrophages and D1 dendritic cells under IFNß-neutralizing conditions. To analyze if all IFNα isoforms are recognized by the intrabody was not in the focus of this study. Provided that binding of the intrabody to all isoforms was confirmed, the establishment of transgenic intrabody mice would be promising for studying the function of IFNα during viral infection and autoimmune diseases.


Asunto(s)
Células Dendríticas/inmunología , Retículo Endoplásmico/inmunología , Interferón-alfa/antagonistas & inhibidores , Macrófagos/inmunología , Anticuerpos de Cadena Única/farmacología , Replicación Viral/efectos de los fármacos , Animales , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Interferón-alfa/efectos de los fármacos , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Células RAW 264.7
6.
PLoS Pathog ; 14(8): e1007174, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30071107

RESUMEN

In the current study, we used a mouse model and human blood samples to determine the effects of chronic alcohol consumption on immune responses during Mycobacterium tuberculosis (Mtb) infection. Alcohol increased the mortality of young mice but not old mice with Mtb infection. CD11b+Ly6G+ cells are the major source of IFN-α in the lungs of Mtb-infected alcohol-fed young mice, and IFN-α enhances macrophage necroptosis in the lungs. Treatment with an anti-IFNAR-1 antibody enhanced the survival of Mtb-infected alcohol-fed young mice. In response to Mtb, peripheral blood mononuclear cells (PBMCs) from alcoholic young healthy individuals with latent tuberculosis infection (LTBI) produced significantly higher amounts of IFN-α than those from non-alcoholic young healthy LTBI+ individuals and alcoholic and non-alcoholic old healthy LTBI+ individuals. Our study demonstrates that alcohol enhances IFN-α production by CD11b+Ly6G+ cells in the lungs of young Mtb-infected mice, which leads to macrophage necroptosis and increased mortality. Our findings also suggest that young alcoholic LTBI+ individuals have a higher risk of developing active TB infection.


Asunto(s)
Consumo de Bebidas Alcohólicas/inmunología , Interferón-alfa/biosíntesis , Interferón-alfa/efectos de los fármacos , Tuberculosis/inmunología , Adulto , Animales , Susceptibilidad a Enfermedades/inmunología , Femenino , Humanos , Interferón-alfa/inmunología , Tuberculosis Latente/inmunología , Masculino , Ratones , Mycobacterium tuberculosis
7.
Clin Immunol ; 197: 19-26, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30056130

RESUMEN

B-cell activating factor (BAFF) has been proposed to play a crucial role in the pathogenesis of chronic rhinosinusitis with nasal polyp (CRSwNP). The aim of this study was to evaluate the role of toll-like receptor (TLR) 9-mediated BAFF activation on the pathogenesis of CRSwNP. NP and uncinate tissue (UT) were obtained from patients with CRSwNP or CRS without NP, and control subjects. The expression of TLR9, high mobility group box-1 protein (HMGB1), type I interferon (IFN), BAFF, and anti-double stranded DNA (dsDNA) antibody were examined in the tissues and the cultured dispersed NP cells (DNPCs). The expression of TLR9, HMGB1, type I IFN, BAFF, and anti-dsDNA antibody were elevated in NP tissue compared to the UTs. Exposure to TLR9 agonist increased the type I IFN expression in vitro, which further increased BAFF production. In conclusion, we provided a novel therapeutic potential of TLR9 agonist in CRSwNP.


Asunto(s)
Factor Activador de Células B/genética , Proteína HMGB1/genética , Pólipos Nasales/metabolismo , Rinitis/metabolismo , Sinusitis/metabolismo , Receptor Toll-Like 9/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Factor Activador de Células B/efectos de los fármacos , Factor Activador de Células B/metabolismo , Enfermedad Crónica , Femenino , Seno Frontal/metabolismo , Proteína HMGB1/metabolismo , Humanos , Técnicas In Vitro , Interferón-alfa/efectos de los fármacos , Interferón-alfa/genética , Interferón-alfa/metabolismo , Interferón beta/efectos de los fármacos , Interferón beta/genética , Interferón beta/metabolismo , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Receptor Toll-Like 9/agonistas
8.
J Acquir Immune Defic Syndr ; 75(5): 588-596, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28692581

RESUMEN

Plasmacytoid dendritic cells (pDCs) play a crucial role in host antiviral immune response through secretion of type I interferon. Interferon alpha (IFNα), a type I IFN, is critical for mounting the initial response to viral pathogens. A consequence of Human Immunodeficiency Virus-1 (HIV) infection is a decrease in both pDC number and function, but prolonged pDC activity has been linked with progression from HIV infection to the development of AIDS. Patients with HIV in the United States routinely use cannabinoid-based therapies to combat the side effects of HIV infection and antiretroviral therapy. However, cannabinoids, including Δ-tetrahydrocannabinol (THC), are well-characterized immunosuppressants. Here, we report that THC suppressed secretion of IFNα by pDC from both healthy and HIV+ donors through a mechanism involving impaired phosphorylation of interferon regulatory factor 7. These results suggest that THC can suppress pDC function during the early host antiviral response by dampening pDC activation.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Dronabinol/farmacología , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Interferón-alfa/efectos de los fármacos , Interferón-alfa/metabolismo , Adulto , Anciano , Células Dendríticas/inmunología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Masculino , Persona de Mediana Edad
9.
Arthritis Rheumatol ; 69(7): 1495-1501, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28426911

RESUMEN

OBJECTIVE: Gain-of-function mutations in TMEM173, encoding the stimulator of interferon (IFN) genes (STING) protein, underlie a novel type I interferonopathy that is minimally responsive to conventional immunosuppressive therapies and associated with high frequency of childhood morbidity and mortality. STING gain-of-function causes constitutive oversecretion of IFN. This study was undertaken to determine the effects of a TANK-binding kinase 1 (TBK-1)/IKKɛ inhibitor (BX795) on secretion and signaling of IFN in primary peripheral blood mononuclear cells (PBMCs) from patients with mutations in STING. METHODS: PBMCs from 4 patients with STING-associated disease were treated with BX795. The effect of BX795 on IFN pathways was assessed by Western blotting and an IFNß reporter assay, as well as by quantification of IFNα in cell lysates, staining for STAT-1 phosphorylation, and measurement of IFN-stimulated gene (ISG) messenger RNA (mRNA) expression. RESULTS: Treatment of PBMCs with BX795 inhibited the phosphorylation of IFN regulatory factor 3 and IFNß promoter activity induced in HEK 293T cells by cyclic GMP-AMP or by genetic activation of STING. In vitro exposure to BX795 inhibited IFNα production in PBMCs of patients with STING-associated disease without affecting cell survival. In addition, BX795 decreased STAT-1 phosphorylation and ISG mRNA expression independent of IFNα blockade. CONCLUSION: These findings demonstrate the effect of BX795 on reducing type I IFN production and IFN signaling in cells from patients with gain-of-function mutations in STING. A combined inhibition of TBK-1 and IKKɛ therefore holds potential for the treatment of patients carrying STING mutations, and may also be relevant in other type I interferonopathies.


Asunto(s)
Factor 3 Regulador del Interferón/efectos de los fármacos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/efectos de los fármacos , Interferón-alfa/efectos de los fármacos , Interferón beta/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Proteínas de la Membrana/efectos de los fármacos , Pirimidinas/farmacología , Tiofenos/farmacología , Western Blotting , Niño , Células HEK293 , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Técnicas In Vitro , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/efectos de los fármacos , Factores Reguladores del Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón-alfa/inmunología , Interferón beta/inmunología , Leucocitos Mononucleares/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Mutación , Nucleótidos Cíclicos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo
10.
Clin Infect Dis ; 64(12): 1686-1695, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28329286

RESUMEN

BACKGROUND.: Treatment with latency reversing agents (LRAs) enhances human immunodeficiency virus type 1 (HIV-1) transcription in vivo but leads to only modest reductions in the size of the reservoir, possibly due to insufficient immune-mediated elimination of infected cells. We hypothesized that a single drug molecule-a novel Toll-like receptor 9 (TLR9) agonist, MGN1703-could function as an enhancer of innate immunity and an LRA in vivo. METHODS.: We conducted a single-arm, open-label study in which 15 virologically suppressed HIV-1-infected individuals on antiretroviral therapy received 60 mg MGN1703 subcutaneously twice weekly for 4 weeks. We characterized plasmacytoid dendritic cell, natural killer (NK), and T-cell activation using flow cytometry on baseline and after 4 weeks of treatment. HIV-1 transcription was quantified by measuring plasma HIV-1 RNA during MGN1703 administration. RESULTS.: In accordance with the cell type-specific expression of TLR9, MGN1703 treatment led to pronounced activation of plasmacytoid dendritic cells and substantial increases in plasma interferon-α2 levels (P < .0001). Consistently, transcription of interferon-stimulated genes (eg, OAS1, ISG15, Mx1; each P < .0001) were upregulated in CD4+ T cells as demonstrated by RNA sequencing. Further, proportions of activated cytotoxic NK cells and CD8+ T cells increased significantly during MGN1703 dosing, suggesting an enhancement of cellular immune responses. In 6 of 15 participants, plasma HIV-1 RNA increased from <20 copies/mL to >1500 copies/mL (range, 21-1571 copies/mL) during treatment. CONCLUSIONS.: TLR9 agonist treatment in HIV infection has a dual potential by increasing HIV-1 transcription and enhancing cytotoxic NK cell activation, both of which are key outcomes in HIV-1 eradication therapy. CLINICAL TRIALS REGISTRATION.: NCT02443935.


Asunto(s)
ADN/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Receptor Toll-Like 9/agonistas , Viremia/tratamiento farmacológico , 2',5'-Oligoadenilato Sintetasa/genética , Terapia Antirretroviral Altamente Activa , Linfocitos T CD8-positivos/efectos de los fármacos , Citocinas/genética , ADN/administración & dosificación , Células Dendríticas/efectos de los fármacos , Femenino , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inmunidad Innata/genética , Interferón-alfa/sangre , Interferón-alfa/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Proteínas de Resistencia a Mixovirus/genética , ARN Viral/efectos adversos , ARN Viral/sangre , Receptor Toll-Like 9/genética , Ubiquitinas/genética , Viremia/sangre , Latencia del Virus/efectos de los fármacos
11.
Vaccine ; 34(47): 5700-5703, 2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27745950

RESUMEN

We recently identified a population of 10% of infants who respond with sub-protective antibody levels to most routine primary pediatric vaccinations due to altered innate and adaptive immune responses. We term these infants as low vaccine responders (LVRs). Here we report new data showing that TLR7/8 agonist - R848 stimulation of PBMCs of LVR infants elicit significantly lower IFN-α, IL-12p70 and IL-1ß, while inducing higher levels of CCL5 (RANTES) compared to normal vaccine responder (NVR) infants.


Asunto(s)
Formación de Anticuerpos/inmunología , Inmunidad Innata , Leucocitos Mononucleares/inmunología , Vacunas/inmunología , Inmunidad Adaptativa , Células Cultivadas , Quimiocina CCL5/sangre , Humanos , Imidazoles/farmacología , Lactante , Interferón-alfa/sangre , Interferón-alfa/efectos de los fármacos , Interleucina-12/antagonistas & inhibidores , Interleucina-12/sangre , Interleucina-1beta/sangre , Interleucina-1beta/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Estudios Longitudinales , Estudios Prospectivos , Estadísticas no Paramétricas , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 8/antagonistas & inhibidores , Vacunación , Vacunas/administración & dosificación
12.
J Biotechnol ; 233: 6-16, 2016 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-27346232

RESUMEN

Type I Interferons (IFNs-I) are species-specific glycoproteins which play an important role as primary defence against viral infections and that can also modulate the adaptive immune system. In some autoimmune diseases, interferons (IFNs) are over-produced. IFNs are widely used as biopharmaceuticals for a variety of cancer indications, chronic viral diseases, and for their immuno-modulatory action in patients with multiple sclerosis; therefore, increasing their therapeutic efficiency and decreasing their side effects is of high clinical value. In this sense, it is interesting to find molecules that can modulate the activity of IFNs. In order to achieve that, it was necessary to establish a simple, fast and robust assay to analyze numerous compounds simultaneously. We developed four reporter gene assays (RGAs) to identify IFN activity modulator compounds by using WISH-Mx2/EGFP, HeLa-Mx2/EGFP, A549-Mx2/EGFP, and HEp2-Mx2/EGFP reporter cell lines (RCLs). All of them present a Z' factor higher than 0.7. By using these RGAs, natural and synthetic compounds were analyzed simultaneously. A total of 442 compounds were studied by the Low Throughput Screening (LTS) assay using the four RCLs to discriminate between their inhibitory or enhancing effects on IFN activity. Some of them were characterized and 15 leads were identified. Finally, one promising candidate with enhancing effect on IFN-α/-ß activity and five compounds with inhibitory effect were described.


Asunto(s)
Descubrimiento de Drogas/métodos , Genes Reporteros/genética , Interferón-alfa/efectos de los fármacos , Interferón-alfa/fisiología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas Genéticas , Células HeLa , Humanos , Reproducibilidad de los Resultados
13.
Arthritis Rheumatol ; 68(11): 2686-2696, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27338297

RESUMEN

OBJECTIVE: Increased expression of type I interferon (IFN) and a broad signature of type I IFN-induced gene transcripts are observed in patients with systemic lupus erythematosus (SLE) and other systemic autoimmune diseases. To identify disease-relevant triggers of the type I IFN pathway, this study sought to investigate whether endogenous virus-like genomic repeat elements, normally silent, are expressed in patients with systemic autoimmune disease, and whether these retroelements could activate an innate immune response and induce type I IFN. METHODS: Expression of type I IFN and long interspersed nuclear element 1 (LINE-1; L1) was studied by polymerase chain reaction, Western blotting, and immunohistochemistry in samples of kidney tissue from patients with lupus nephritis and minor salivary gland (MSG) tissue from patients with primary Sjögren's syndrome (SS). Induction of type I IFN by L1 was investigated by transfection of plasmacytoid dendritic cells (PDCs) or monocytes with an L1-encoding plasmid or L1 RNA. Involvement of innate immune pathways and altered L1 methylation were assessed. RESULTS: Levels of L1 messenger RNA transcripts were increased in lupus nephritis kidneys and in MSG tissue from patients with SS. Transcript expression correlated with the expression of type I IFN and L1 DNA demethylation. L1 open-reading frame 1/p40 protein and IFNß were expressed in MSG ductal epithelial cells and in lupus nephritis kidneys, and IFNα was detected in infiltrating PDCs. Transfection of PDCs or monocytes with L1-encoding DNA or RNA induced type I IFN. Inhibition of Toll-like receptor 7 (TLR-7)/TLR-8 reduced the induction of IFNα by L1 in PDCs, and an inhibitor of IKKε/TANK-binding kinase 1 abrogated the induction of type I IFN by L1 RNA in monocytes. CONCLUSION: L1 genomic repeat elements represent endogenous nucleic acid triggers of the type I IFN pathway in SLE and SS and may contribute to initiation or amplification of autoimmune disease.


Asunto(s)
Interferón Tipo I/inmunología , Elementos de Nucleótido Esparcido Largo/genética , Lupus Eritematoso Sistémico/genética , Síndrome de Sjögren/genética , Adulto , Anciano , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Western Blotting , Metilación de ADN , Células Dendríticas/inmunología , Femenino , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Inmunidad Innata/inmunología , Inmunohistoquímica , Interferón Tipo I/efectos de los fármacos , Interferón Tipo I/metabolismo , Interferón-alfa/efectos de los fármacos , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Interferón beta/efectos de los fármacos , Interferón beta/inmunología , Interferón beta/metabolismo , Riñón/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Elementos de Nucleótido Esparcido Largo/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Glándulas Salivales Menores/metabolismo , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/metabolismo , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 8/antagonistas & inhibidores , Receptor Toll-Like 9/antagonistas & inhibidores
14.
BMC Gastroenterol ; 16: 40, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27005303

RESUMEN

BACKGROUND: Breastfeeding is associated with a variety of positive health outcomes in children and is recommended exclusively for the first 6 months of life; however, 50-70 % of infants in the US are formula-fed. To test the hypothesis that immune system development and function in neonates and infants are significantly influenced by diet, 2-day old piglets were fed soy or milk formula (n = 6/group/gender) until day 21 and compared to a sow-fed group (n = 6/gender). METHODS: Histomorphometric analyses of ileum, jejunum and Peyer's patches were carried out, to determine the inflammation status, mRNA and protein expression of pro-inflammatory, anti-inflammatory and growth-related chemokines and cytokines. RESULTS: In formula-fed animals, increases in ileum and jejunum villus height and crypt depth were observed in comparison to sow-fed animals (jejunum, p < 0.01 villus height, p < 0.04 crypt depth; ileum p < 0.001 villus height, p < 0.002 crypt depth). In formula-fed the lymphoid follicle size (p < 0.01) and germinal centers (p < 0.01) with in the Peyer's patch were significantly decreased in comparison to sow-fed, indicating less immune education. In ileum, formula diet induced significant up-regulation of AMCFII, IL-8, IL-15, VEGFA, LIF, FASL, CXCL11, CCL4, CCL25 and down-regulation of IL-6, IL-9, IL-10, IL-27, IFNA4, CSF3, LOC100152038, and LOC100736831 at the transcript level. We have confirmed some of the mRNA data by measuring protein, and significant down-regulation of anti-inflammatory molecule IL-10 in comparison to sow-fed piglets was observed. To further determine the membrane protein expression in the ileum, VE-cadherin, occludin, and claudin-3, Western blot analyses were conducted. Sow fed piglets showed significantly more VE-Cadherin, which associated with levels of calcium, and putrescine measured. It is possible that differences in GI tract and immune development are related to shifts in the microbiome; notably, there were 5-fold higher amounts of Lactobacillaceae spp and 3 fold higher Clostridia spp in the sow fed group in comparison to milk formula-fed piglets, whereas in milk formula-fed pigs Enterobacteriaceae spp was 5-fold higher. CONCLUSION: In conclusion, formula diet alters GI morphology, microbial abundance, intestinal barrier protein VE-cadherin and anti-inflammatory molecule IL-10 expression. Further characterization of formula effects could lead to modification of infant formula to improve immune function, reduce inflammation and prevent conditions such as allergies and infections.


Asunto(s)
Antígenos CD/genética , Cadherinas/genética , Citocinas/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Fórmulas Infantiles/farmacología , Intestino Delgado/efectos de los fármacos , Leche , ARN Mensajero/efectos de los fármacos , Alimentos de Soja , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Cadherinas/metabolismo , Calcio/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dieta , Regulación hacia Abajo , Proteína Ligando Fas/efectos de los fármacos , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Humanos , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/microbiología , Íleon/patología , Recién Nacido , Interferón-alfa/efectos de los fármacos , Interferón-alfa/genética , Interferón-alfa/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-15/genética , Interleucina-15/metabolismo , Interleucina-27/genética , Interleucina-27/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/efectos de los fármacos , Interleucina-8/genética , Interleucina-8/metabolismo , Interleucina-9/genética , Interleucina-9/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestino Delgado/patología , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/microbiología , Yeyuno/patología , Factor Inhibidor de Leucemia/efectos de los fármacos , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Ganglios Linfáticos Agregados/efectos de los fármacos , Ganglios Linfáticos Agregados/inmunología , ARN Mensajero/metabolismo , Porcinos , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
15.
Antiviral Res ; 126: 69-80, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26738783

RESUMEN

Influenza A viruses (IAVs) impact the public health and global economy by causing yearly epidemics and occasional pandemics. Several anti-IAV drugs are available and many are in development. However, the question remains which of these antiviral agents may allow activation of immune responses and protect patients against co- and re-infections. To answer to this question, we analysed immuno-modulating properties of the antivirals saliphenylhalamide (SaliPhe), SNS-032, obatoclax, and gemcitabine, and found that only gemcitabine did not impair immune responses in infected cells. It also allowed activation of innate immune responses in lipopolysaccharide (LPS)- and interferon alpha (IFNα)-stimulated macrophages. Moreover, immuno-mediators produced by gemcitabine-treated IAV-infected macrophages were able to prime immune responses in non-infected cells. Thus, we identified an antiviral agent which might be beneficial for treatment of patients with severe viral infections.


Asunto(s)
Antineoplásicos/farmacología , Antivirales/farmacología , Factores Inmunológicos/farmacología , Gripe Humana/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Macrófagos/virología , Amidas/farmacología , Células Cultivadas , Coinfección/tratamiento farmacológico , Coinfección/virología , Citocinas/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Humanos , Inmunidad Innata/efectos de los fármacos , Indoles , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Gripe Humana/virología , Interferón-alfa/efectos de los fármacos , Interferón-alfa/inmunología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Oxazoles/farmacología , Fosfoproteínas/metabolismo , Pirroles/farmacología , ARN Viral/biosíntesis , Salicilatos/farmacología , Tiazoles/farmacología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología , Gemcitabina
16.
J Biol Chem ; 290(24): 14979-85, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25925951

RESUMEN

We report the three-dimensional structure of human interferon α-2A (IFN-α2A) bound to the Fab fragment of a therapeutic monoclonal antibody (sifalimumab; IgG1/κ). The structure of the corresponding complex was solved at a resolution of 3.0 Å using molecular replacement and constitutes the first reported structure of a human type I IFN bound to a therapeutic antibody. This study revealed the major contribution made by the first complementarity-determining region in each of sifalimumab light and heavy chains. These data also provided the molecular basis for sifalimumab mechanism of action. We propose that its interferon-neutralizing properties are the result of direct competition for IFN-α2A binding to the IFN receptor subunit 1 (IFNAR1) and do not involve inhibiting IFN-α2A binding to the IFN receptor subunit 2 (IFNAR2).


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Interferón-alfa/efectos de los fármacos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Cristalografía por Rayos X , Interferón alfa-2 , Interferón-alfa/inmunología , Modelos Moleculares , Conformación Proteica , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/inmunología
19.
J Oral Pathol Med ; 43(9): 711-21, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24762323

RESUMEN

The mevalonate pathway (MVP) and the anti-angiogenic effect of bisphosphonates have been shown to play a role in the pathogenesis of bisphosphonate-related osteonecrosis of the jaw (BRONJ). This study determined the effect of the bisphosphonate, zoledronic acid and the replenishment of the MVP by geranylgeraniol on human gingival fibroblasts. Cell viability, apoptosis, morphological analysis using transmission electron microscopy, and gene expression for vascular endothelial growth factor A, bone morphogenic protein 2, ras homologue gene family member B, epiregulin and interferon-alpha were conducted. Results showed cellular viability was decreased in the presence of zoledronic acid and the co-addition of zoledronic acid with geranylgeraniol restored cell viability to control levels. Caspase 3/7 was detected in zoledronic-acid-treated cells indicating apoptosis. Transmission electron microscopy revealed dilation of the rough endoplasmic reticulum with zoledronic acid and the appearance of multiple lipid-like vesicles following the addition of geranylgeraniol. Zoledronic acid significantly (P < 0.05, FR > ± 2) up-regulated vascular endothelial growth factor A, bone morphogenic protein 2, ras homologue gene family member B and epiregulin at one or more time points but not interferon-alpha. Addition of geranylgeraniol resulted in a reduction in the expression of all five genes compared with zoledronic-acid-treated human gingival fibroblasts. The study concluded geranylgeraniol partially reversed the effects of zoledronic acid in human gingival fibroblasts both at the cellular and genetic levels, suggesting the regulation of these genes is mediated via the mevalonate pathway.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Difosfonatos/farmacología , Diterpenos/farmacología , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Imidazoles/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Adulto , Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 2/efectos de los fármacos , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Epirregulina/análisis , Farnesol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Encía/citología , Humanos , Interferón-alfa/efectos de los fármacos , Ácido Mevalónico/metabolismo , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Neovascularización Fisiológica/genética , Fosfatos de Poliisoprenilo/metabolismo , Sesquiterpenos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Ácido Zoledrónico , Proteína de Unión al GTP rhoB/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...