Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 11(28): e2401772, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38767114

RESUMEN

Polycystic ovary syndrome (PCOS) is associated with a low-grade inflammation, but it is unknown how hyperandrogenism, the hallmark of PCOS, affects the immune system. Using a PCOS-like mouse model, it is demonstrated that hyperandrogenism affects immune cell populations in reproductive, metabolic, and immunological tissues differently in a site-specific manner. Co-treatment with an androgen receptor antagonist prevents most of these alterations, demonstrating that these effects are mediated through androgen receptor activation. Dihydrotestosterone (DHT)-exposed mice displayed a drastically reduced eosinophil population in the uterus and visceral adipose tissue (VAT). A higher frequency of natural killer (NK) cells and elevated levels of IFN-γ and TNF-α are seen in uteri of androgen-exposed mice, while NK cells in VAT and spleen displayed a higher expression level of CD69, a marker of activation or tissue residency. Distinct alterations of macrophages in the uterus, ovaries, and VAT are also found in DHT-exposed mice and can potentially be linked to PCOS-like traits of the model. Indeed, androgen-exposed mice are insulin-resistant, albeit unaltered fat mass. Collectively, it is demonstrated that hyperandrogenism causes tissue-specific alterations of immune cells in reproductive organs and VAT, which can have considerable implications on tissue function and contribute to the reduced fertility and metabolic comorbidities associated with PCOS.


Asunto(s)
Andrógenos , Modelos Animales de Enfermedad , Síndrome del Ovario Poliquístico , Animales , Síndrome del Ovario Poliquístico/inmunología , Síndrome del Ovario Poliquístico/metabolismo , Femenino , Ratones , Andrógenos/metabolismo , Ratones Endogámicos C57BL , Hiperandrogenismo/inmunología , Hiperandrogenismo/metabolismo
2.
bioRxiv ; 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37986968

RESUMEN

There is currently a lack of tools capable of perturbing genes in both a precise and spatiotemporal fashion. CRISPR's ease of use and flexibility, coupled with light's unparalleled spatiotemporal resolution deliverable from a controllable source, makes optogenetic CRISPR a well-suited solution for precise spatiotemporal gene perturbations. Here we present a new optogenetic CRISPR tool, BLU-VIPR, that diverges from prevailing split-Cas design strategies and instead focuses on optogenetic regulation of gRNA production. This simplifies spatiotemporal gene perturbation and works in vivo with cells previously intractable to optogenetic gene editing. We engineered BLU-VIPR around a new potent blue-light activated transcription factor and ribozyme-flanked gRNA. The BLU-VIPR design is genetically encoded and ensures precise excision of multiple gRNAs from a single mRNA transcript, allowing for optogenetic gene editing in T lymphocytes in vivo.

3.
EMBO Rep ; 24(1): e54944, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36341538

RESUMEN

Melanoma tumors are highly metastatic partly due to the ability of melanoma cells to transition between invasive and proliferative states. However, the mechanisms underlying this plasticity are still not fully understood. To identify new epigenetic regulators of melanoma plasticity, we combined data mining, tumor models, proximity proteomics, and CUT&RUN sequencing. We focus on the druggable family of bromodomain epigenetic readers and identify TRIM28 as a new regulator of melanoma plasticity. We find that TRIM28 promotes the expression of pro-invasive genes and that TRIM28 controls the balance between invasiveness and growth of melanoma cells. We demonstrate that TRIM28 acts via the transcription factor JUNB that directly regulates the expression of pro-invasive and pro-growth genes. Mechanistically, TRIM28 controls the expression of JUNB by negatively regulating its transcriptional elongation by RNA polymerase II. In conclusion, our results demonstrate that a TRIM28-JUNB axis controls the balance between invasiveness and growth in melanoma tumors and suggest that the bromodomain protein TRIM28 could be targeted to reduce tumor spread.


Asunto(s)
Regulación de la Expresión Génica , Melanoma , Humanos , Línea Celular Tumoral , Proteína 28 que Contiene Motivos Tripartito/genética , Melanoma/genética
4.
Nat Commun ; 13(1): 4983, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36127330

RESUMEN

Diabetes is known to increase susceptibility to infections, partly due to impaired granulocyte function and changes in the innate immunity. Here, we investigate the effect of diabetes, and high glucose on the expression of the antimicrobial peptide, psoriasin and the putative consequences for E. coli urinary tract infection. Blood, urine, and urine exfoliated cells from patients are studied. The influence of glucose and insulin is examined during hyperglycemic clamps in individuals with prediabetes and in euglycemic hyperinsulinemic clamped patients with type 1 diabetes. Important findings are confirmed in vivo in type 2 diabetic mice and verified in human uroepithelial cell lines. High glucose concentrations induce lower psoriasin levels and impair epithelial barrier function together with altering cell membrane proteins and cytoskeletal elements, resulting in increasing bacterial burden. Estradiol treatment restores the cellular function with increasing psoriasin and bacterial killing in uroepithelial cells, confirming its importance during urinary tract infection in hyperglycemia. In conclusion, our findings present the effects and underlying mechanisms of high glucose compromising innate immunity.


Asunto(s)
Diabetes Mellitus Experimental , Infecciones por Escherichia coli , Infecciones Urinarias , Animales , Péptidos Antimicrobianos , Escherichia coli/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico , Estradiol/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteína A7 de Unión a Calcio de la Familia S100/metabolismo , Vejiga Urinaria/metabolismo
5.
Biomolecules ; 12(6)2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35740904

RESUMEN

Macrophages are key inflammatory immune cells that display dynamic phenotypes and functions in response to their local microenvironment. In different conditions, macrophage polarization can be induced by high-mobility group box 1 (HMGB1), a nuclear DNA-binding protein that activates innate immunity via the Toll-like receptor (TLR) 4, the receptor for advanced glycation end products (RAGE), and C-X-C chemokine receptor (CXCR) 4. This study investigated the phenotypes of murine bone-marrow-derived macrophages (BMDMs) stimulated with different HMGB1 redox isoforms using bulk RNA sequencing (RNA-Seq). Disulfide HMGB1 (dsHMGB1)-stimulated BMDMs showed a similar but distinct transcriptomic profile to LPS/IFNγ- and LPS-stimulated BMDMs. Fully reduced HMGB1 (frHMGB1) did not induce any significant transcriptomic change. Interestingly, compared to LPS/IFNγ- and LPS-, dsHMGB1-stimulated BMDMs showed lipid metabolism and foam cell differentiation gene set enrichment, and oil red O staining revealed that both dsHMGB1 and frHMGB1 alleviated oxidized low-density lipoprotein (oxLDL)-induced foam cells formation. Overall, this work, for the first time, used transcriptomic analysis by RNA-Seq to investigate the impact of HMGB1 stimulation on BMDM polarization. Our results demonstrated that dsHMGB1 and frHMGB1 induced distinct BMDM polarization phenotypes compared to LPS/IFNγ- and LPS- induced phenotypes.


Asunto(s)
Proteína HMGB1 , Activación de Macrófagos , Transcriptoma , Animales , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones
6.
Ann Rheum Dis ; 81(8): 1151-1161, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35470161

RESUMEN

OBJECTIVE: Neonatal lupus erythematosus (NLE) may develop after transplacental transfer of maternal autoantibodies with cardiac manifestations (congenital heart block, CHB) including atrioventricular block, atrial and ventricular arrhythmias, and cardiomyopathies. The association with anti-Ro/SSA antibodies is well established, but a recurrence rate of only 12%-16% despite persisting maternal autoantibodies suggests that additional factors are required for CHB development. Here, we identify fetal genetic variants conferring risk of CHB and elucidate their effects on cardiac function. METHODS: A genome-wide association study was performed in families with at least one case of CHB. Gene expression was analysed by microarrays, RNA sequencing and PCR and protein expression by western blot, immunohistochemistry, immunofluorescence and flow cytometry. Calcium regulation and connectivity were analysed in primary cardiomyocytes and cells induced from pleuripotent stem cells. Fetal heart performance was analysed by Doppler/echocardiography. RESULTS: We identified DNAJC6 as a novel fetal susceptibility gene, with decreased cardiac expression of DNAJC6 associated with the disease risk genotype. We further demonstrate that fetal cardiomyocytes deficient in auxilin, the protein encoded by DNAJC6, have abnormal connectivity and Ca2+ homoeostasis in culture, as well as decreased cell surface expression of the Cav1.3 calcium channel. Doppler echocardiography of auxilin-deficient fetal mice revealed cardiac NLE abnormalities in utero, including abnormal heart rhythm with atrial and ventricular ectopias, as well as a prolonged atrioventricular time intervals. CONCLUSIONS: Our study identifies auxilin as the first genetic susceptibility factor in NLE modulating cardiac function, opening new avenues for the development of screening and therapeutic strategies in CHB.


Asunto(s)
Bloqueo Atrioventricular , Auxilinas , Animales , Anticuerpos Antinucleares , Bloqueo Atrioventricular/genética , Autoanticuerpos , Corazón Fetal , Estudio de Asociación del Genoma Completo , Bloqueo Cardíaco/congénito , Lupus Eritematoso Sistémico/congénito , Ratones
7.
Gac Med Mex ; 156(4): 334-338, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32831328

RESUMEN

COVID-19 arrived in Latin America early in March 2020. Currently, strategies are being developed in Colombia focusing on the quarantine and social and economic capital reactivation, whereby the expected results are not being obtained. In this article, we propose to review scientific evidence-based literature where information on the operation and adaptation of health systems, and social, economic and solidarity sectors of Colombia is presented. The purpose is to identify COVID-19 implications in the network that provides health services, quality of life and health-disease prognosis in the country, which is not prepared to face crises of social nature and of health systems, as well as the economic and solidarity impacts that are brought about by pandemics and crude episodes of disease.


COVID-19 llegó a Latinoamérica a principios de marzo de 2020. Actualmente, en Colombia se desarrollan estrategias enfocadas en la cuarentena y la reactivación del capital social y económico, con las cuales no se están obteniendo los resultados esperados. En este artículo se propone revisar literatura basada en evidencia científica en la que se exponga información del funcionamiento y adaptación de los sistemas de salud, sectores sociales, económicos y solidarios de Colombia. El objetivo es identificar las implicaciones de COVID-19 en la red prestadora de servicios de salud, calidad de vida, pronóstico de salud-enfermedad en el país, el cual no está preparado para afrontar crisis de orden social, de sistemas de salud e impactos económicos y solidarios que conllevan las pandemias y episodios graves de enfermedad.


Asunto(s)
Infecciones por Coronavirus/epidemiología , Atención a la Salud/organización & administración , Neumonía Viral/epidemiología , Salud Pública , Betacoronavirus/aislamiento & purificación , COVID-19 , Colombia/epidemiología , Humanos , Pandemias , Calidad de Vida , SARS-CoV-2
8.
Gac. méd. Méx ; 156(4): 330-334, Jul.-Aug. 2020.
Artículo en Inglés | LILACS | ID: biblio-1249920

RESUMEN

Abstract COVID-19 arrived to Latin America early in March 2020. Currently, strategies are being developed in Colombia focusing on the quarantine and social and economic capital reactivation, whereby the expected results are not being obtained. In this article, we propose to review scientific evidence-based literature where information on the operation and adaptation of health systems, and social, economic and solidarity sectors of Colombia is presented. The purpose is to identify COVID-19 implications in the network that provides health services, quality of life and health-disease prognosis in the country, which is not prepared to face crises of social nature and of health systems, as well as the economic and solidarity impacts that are brought about by pandemics and crude episodes of disease.


Resumen COVID-19 llegó a Latinoamérica a principios de marzo de 2020. Actualmente, en Colombia se desarrollan estrategias enfocadas en la cuarentena y la reactivación del capital social y económico, con las cuales no se están obteniendo los resultados esperados. En este artículo se propone revisar literatura basada en evidencia científica en la que se exponga información del funcionamiento y adaptación de los sistemas de salud, sectores sociales, económicos y solidarios de Colombia. El objetivo es identificar las implicaciones de COVID-19 en la red prestadora de servicios de salud, calidad de vida, pronóstico de salud-enfermedad en el país, el cual no está preparado para afrontar crisis de orden social, de sistemas de salud e impactos económicos y solidarios que conllevan las pandemias y episodios graves de enfermedad.


Asunto(s)
Humanos , Neumonía Viral/epidemiología , Salud Pública , Infecciones por Coronavirus/epidemiología , Atención a la Salud/organización & administración , Calidad de Vida , Colombia/epidemiología , Pandemias , Betacoronavirus/aislamiento & purificación , SARS-CoV-2 , COVID-19
9.
RMD Open ; 6(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31958275

RESUMEN

OBJECTIVE: In utero exposure of the fetus to Ro/La autoantibodies may lead to congenital heart block (CHB). In the mother, these autoantibodies are associated with activation of the type I interferon (IFN)-system. As maternal autoantibodies are transferred to the fetus during pregnancy, we investigated whether the type I IFN-system is activated also in newborns of anti-Ro/La positive mothers, and whether fetal IFN activation is affected by maternal immunomodulatory treatment. METHODS: Blood drawn at birth from anti-Ro/La positive mothers, their newborns and healthy control pairs was separated into plasma and peripheral blood mononuclear cells (PBMC). PBMC were analysed directly or cultured. mRNA expression was analysed by microarrays, cell surface markers by flow cytometry, and IFNα levels by immunoassays. RESULTS: We observed increased expression of IFN-regulated genes and elevated plasma IFNα levels not only in anti-Ro/La positive women, but also in their newborns. CD14+ monocytes of both anti-Ro/La positive mothers and their neonates showed increased expression of Sialic acid-binding Ig-like lectin-1, indicating cellular activation. Notably, the IFN score of neonates born to mothers receiving immunomodulatory treatment was similar to that of controls, despite persistent IFN activation in the mothers. In both maternal and neonatal PBMC, IFNα production was induced when cells were cultured with anti-Ro/La positive plasma. CONCLUSIONS: Ro/La autoantibody-exposed neonates at risk of CHB have signs of an activated immune system with an IFN signature. This study further demonstrates that neonatal cells can produce IFNα when exposed to autoantibody-containing plasma, and that maternal immunomodulatory treatment may diminish the expression of IFN-regulated genes in the fetus.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Bloqueo Cardíaco/congénito , Interferón Tipo I/inmunología , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Estudios de Casos y Controles , Ecocardiografía Doppler , Femenino , Bloqueo Cardíaco/sangre , Bloqueo Cardíaco/embriología , Bloqueo Cardíaco/inmunología , Humanos , Recién Nacido , Interferón Tipo I/sangre , Masculino , Intercambio Materno-Fetal/inmunología , Embarazo , Complicaciones del Embarazo/inmunología , Enfermedades Reumáticas/inmunología , Suecia , Transcriptoma , Adulto Joven
10.
Immunology ; 159(3): 335-343, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31755557

RESUMEN

TRIM21 is an interferon-stimulated E3 ligase that controls the activity of pattern-recognition signaling via ubiquitination of interferon regulatory factors and DDX41. Previous studies on the role of TRIM21 in innate immune responses have yielded contradictory results, suggesting that the role of TRIM21 is cell specific. Here, we report that bone-marrow-derived macrophages (BMDMs) generated from Trim21-/- mice have reduced expression of mature macrophage markers. Reflecting their reduced differentiation in response to macrophage colony-stimulating factor (M-CSF), Trim21-/- BMDMs had decreased expression of M-CSF signature genes. Although Trim21-/- BMDMs responded normally to Toll-like receptor 9 (TLR9) activation, they produced lower levels of pro-inflammatory cytokines in response to the TLR2 agonist PAM3CSK4. In line with this, the response to infection with the Bacillus Calmette-Guérin strain of Mycobacterium bovis was also diminished in Trim21-/- BMDMs. Our results indicate that TRIM21 controls responses to TLR2 agonists.


Asunto(s)
Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Ribonucleoproteínas/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Interacciones Huésped-Patógeno , Lipopéptidos/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/inmunología , Mycobacterium bovis/patogenicidad , Fenotipo , Ribonucleoproteínas/deficiencia , Ribonucleoproteínas/genética , Transducción de Señal , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/genética
11.
J Biol Chem ; 294(30): 11404-11419, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31160341

RESUMEN

The E3 ubiquitin-protein ligase TRIM21, of the RING-containing tripartite motif (TRIM) protein family, is a major autoantigen in autoimmune diseases and a modulator of innate immune signaling. Together with ubiquitin-conjugating enzyme E2 E1 (UBE2E1), TRIM21 acts both as an E3 ligase and as a substrate in autoubiquitination. We here report a 2.82-Å crystal structure of the human TRIM21 RING domain in complex with the human E2-conjugating UBE2E1 enzyme, in which a ubiquitin-targeted TRIM21 substrate lysine was captured in the UBE2E1 active site. The structure revealed that the direction of lysine entry is similar to that described for human proliferating cell nuclear antigen (PCNA), a small ubiquitin-like modifier (SUMO)-targeted substrate, and thus differs from the canonical SUMO-targeted substrate entry. In agreement, we found that critical UBE2E1 residues involved in the capture of the TRIM21 substrate lysine are conserved in ubiquitin-conjugating E2s, whereas residues critical for SUMOylation are not conserved. We noted that coordination of the acceptor lysine leads to remodeling of amino acid side-chain interactions between the UBE2E1 active site and the E2-E3 direct interface, including the so-called "linchpin" residue conserved in RING E3s and required for ubiquitination. The findings of our work support the notion that substrate lysine activation of an E2-E3-connecting allosteric path may trigger catalytic activity and contribute to the understanding of specific lysine targeting by ubiquitin-conjugating E2s.


Asunto(s)
Lisina/metabolismo , Ribonucleoproteínas/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Estructura Molecular , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ribonucleoproteínas/química , Alineación de Secuencia , Especificidad por Sustrato , Enzimas Ubiquitina-Conjugadoras/química
12.
Eur J Immunol ; 49(2): 313-322, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30307034

RESUMEN

Systemic autoimmune diseases are characterized by the overexpression of type I IFN stimulated genes, and accumulating evidence indicate a role for type I IFNs in these diseases. However, the underlying mechanisms for this are still poorly understood. To explore the role of type I IFN regulated miRNAs in systemic autoimmune disease, we characterized cellular expression of miRNAs during both acute and chronic type I IFN responses. We identified a T cell-specific reduction of miR-31-5p levels, both after intramuscular injection of IFNß and in patients with Sjögren's syndrome (SjS). To interrogate the role of miR-31-51p in T cells we transfected human CD4+ T cells with a miR-31-5p inhibitor and performed metabolic measurements. This identified an increase in basal levels of glucose metabolism after inhibition of miR-31-5p. Furthermore, treatment with IFN-α also increased the basal levels of human CD4+ T-cell metabolism. In all, our results suggest that reduced levels of miR-31-5p in T cells of SjS patients support autoimmune T-cell responses during chronic type I IFN exposure.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Metabolismo Energético/inmunología , MicroARNs/inmunología , Síndrome de Sjögren/inmunología , Linfocitos T CD4-Positivos/patología , Metabolismo Energético/efectos de los fármacos , Femenino , Humanos , Interferón-alfa/inmunología , Interferón-alfa/farmacología , Interferón beta/inmunología , Interferón beta/farmacología , Masculino , Síndrome de Sjögren/patología
13.
Nat Commun ; 9(1): 4845, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451869

RESUMEN

Circulating monocytes can compete for virtually any tissue macrophage niche and become long-lived replacements that are phenotypically indistinguishable from their embryonic counterparts. As the factors regulating this process are incompletely understood, we studied niche competition in the brain by depleting microglia with >95% efficiency using Cx3cr1CreER/+R26DTA/+ mice and monitored long-term repopulation. Here we show that the microglial niche is repopulated within weeks by a combination of local proliferation of CX3CR1+F4/80lowClec12a- microglia and infiltration of CX3CR1+F4/80hiClec12a+ macrophages that arise directly from Ly6Chi monocytes. This colonization is independent of blood brain barrier breakdown, paralleled by vascular activation, and regulated by type I interferon. Ly6Chi monocytes upregulate microglia gene expression and adopt microglia DNA methylation signatures, but retain a distinct gene signature from proliferating microglia, displaying altered surface marker expression, phagocytic capacity and cytokine production. Our results demonstrate that monocytes are imprinted by the CNS microenvironment but remain transcriptionally, epigenetically and functionally distinct.


Asunto(s)
Encéfalo/inmunología , Linaje de la Célula/inmunología , Regulación de la Expresión Génica/inmunología , Microglía/inmunología , Monocitos/inmunología , Traslado Adoptivo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos Ly/genética , Antígenos Ly/inmunología , Proteínas Bacterianas/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Encéfalo/citología , Encéfalo/efectos de la radiación , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Linaje de la Célula/efectos de la radiación , Proliferación Celular , Metilación de ADN , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Proteínas Luminiscentes/inmunología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/efectos de la radiación , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/efectos de la radiación , Monocitos/citología , Monocitos/efectos de la radiación , Monocitos/trasplante , Fagocitosis , Receptores Mitogénicos/genética , Receptores Mitogénicos/inmunología , Transducción de Señal , Quimera por Trasplante , Irradiación Corporal Total
15.
Exp Dermatol ; 26(8): 713-720, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27943421

RESUMEN

Tripartite motif-containing protein 21 (TRIM21) regulates pro-inflammatory cytokines and type I interferons and acts as an autoantigen in certain autoimmune diseases, but TRIM21 has not been investigated in psoriasis. It has been suggested that TRIM21 may have a dual function; in the early phase of inflammation, it may function as a stimulator; but upon immune stimulation, its ubiquitinating mode of action may shift from stabilization to degradation of IRF3 causing inhibition of the immune responses. The imiquimod (IMQ)-induced psoriasis-like mouse model displays features similar to those of human psoriasis. However, chronicity is lacking in this model. We investigated whether the role of TRIM21 in psoriasis was pro-inflammatory or anti-inflammatory. We hypothesized that a shift of the TRIM21-ubiquitinating mode of action may explain the lack of chronicity in the IMQ-induced psoriasis-like mouse model. We showed that TRIM21 expression is increased in lesional psoriatic skin and in the early phase of IMQ-induced inflammation both in vitro and in vivo. Surprisingly, inflammation was significantly less pronounced in TRIM21 knockout mice than in wild-type mice as shown by ear thickness measured at days 8, 9 and 10 after treatment start, by spleen weight as a marker of systemic effect of IMQ at 10 days after treatment start and by expression of IL-12p40 at days 3 and 10 after treatment start and IL-17A at day 3 after treatment start. Therefore, induction of TRIM21 expression cannot explain the lack of chronicity in the IMQ-induced psoriasis-like skin inflammation mouse model.


Asunto(s)
Aminoquinolinas/efectos adversos , Antineoplásicos/efectos adversos , Psoriasis/inducido químicamente , Ribonucleoproteínas/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Imiquimod , Ratones Noqueados , Psoriasis/metabolismo , Ribonucleoproteínas/genética
16.
Biochem Biophys Res Commun ; 473(4): 789-794, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27003259

RESUMEN

Endoplasmic reticulum (ER) stress is a physiological response to protein overload or misfolded proteins in the ER. Certain anti-cancer drugs, e.g. bortezomib and nelfinavir, induce ER stress implying that this could be a successful therapeutic strategy against several forms of cancer. To find novel ER-stress inducers we screened a panel of natural and synthetic Toll-like receptor (TLR) agonists against human keratinocytes and identified the anti-cancer drug imiquimod (IMQ) as a potent inducer of ER stress. Other TLR7 and TLR8 agonists, including resiquimod and gardiquimod, did not induce ER stress, demonstrating that IMQ induces ER stress independently of TLR7 and TLR8. We further confirmed this by showing that IMQ could still induce ER stress in mouse Tlr7(-/-) cells. IMQ also induced a rapid and transient influx of extracellular Ca(2+) together with the release of Ca(2+) from internal stores. Depletion of Ca(2+) from the ER is a known cause of ER stress suggesting that IMQ induces ER stress via depletion of ER Ca(2+). The ER-stress inducing property of IMQ is possibly of importance for its efficacy in treating basal cell carcinoma, in situ melanoma, and squamous cell carcinoma. Our data could potentially be harnessed for rational design of even more potent ER-stress inducers and new anti-cancer drugs.


Asunto(s)
Aminoquinolinas/farmacología , Calcio/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Queratinocitos/fisiología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Animales , Antineoplásicos/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Imiquimod , Queratinocitos/efectos de los fármacos , Ratones
17.
J Immunol ; 196(1): 91-6, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26590315

RESUMEN

Patients with systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS) are typically characterized by the presence of autoantibodies and an IFN-signature. The strength of the IFN-signature positively correlates with disease severity, suggesting that type I IFNs are active players in these diseases. BAFF is a cytokine critical for development and proper selection of B cells, and the targeting of BAFF has emerged as a successful treatment strategy of SLE. Previous reports have suggested that BAFF expression is directly induced by type I IFNs, but the precise mechanism for this remains unknown. In this article, we demonstrate that BAFF is a bona fide ISG and that IFN regulatory factors (IRFs) control the expression of BAFF. We identify IRF1 and IRF2 as positive regulators of BAFF transcription and IRF4 and IRF8 as potent repressors; in addition, we have mapped the precise binding site for these factors in the BAFF promoter. IFN-ß injections induced BAFF expression mainly in neutrophils and monocytes, and BAFF expression in neutrophils from pSS patients strongly correlated with the strength of the IFN-signature. In summary, we show that BAFF expression is directly induced by type I IFNs via IRF1 and IRF2, whereas IRF4 and IRF8 are negative regulators of BAFF expression. These data suggest that type I IFN blockade in SLE and pSS patients will lead to downregulation of BAFF and a consequential reduction of autoreactive B cell clones and autoantibodies.


Asunto(s)
Factor Activador de Células B/inmunología , Factor 1 Regulador del Interferón/inmunología , Factor 2 Regulador del Interferón/inmunología , Factores Reguladores del Interferón/inmunología , Interferón beta/inmunología , Autoanticuerpos/inmunología , Factor Activador de Células B/sangre , Factor Activador de Células B/genética , Linfocitos B/inmunología , Secuencia de Bases , Sitios de Unión/genética , Células Cultivadas , Femenino , Humanos , Lupus Eritematoso Sistémico/inmunología , Masculino , Datos de Secuencia Molecular , Monocitos/inmunología , Esclerosis Múltiple/inmunología , Neutrófilos/inmunología , Síndrome de Sjögren/inmunología , Transcripción Genética/genética
18.
J Immunol ; 191(7): 3753-63, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23975864

RESUMEN

Tripartite-motif 21 (TRIM21) is an E3 ubiquitin ligase that regulates innate immune responses by ubiquitinating IFN regulatory factors (IRFs). TRIM21 is mainly found in hematopoietic cells in which its expression is induced by IFNs during viral. infections and in systemic autoimmune diseases such as systemic lupus erythematosus and Sjögren's syndrome. However, the exact molecular mechanism by which the expression of the Trim21 gene is regulated is unknown. In this study, we demonstrate that IFNs induce Trim21 expression in immune cells via IRFs and that IFN-α and IFN-ß are the most potent inducers of Trim21. A functional IFN-stimulated response element but no conserved IFN-γ-activated site was detected in the promoter of Trim21. IRF1 and IRF2 strongly induced Trim21 expression in an IFN-stimulated response element-dependent manner, whereas IRF4 and IRF8 strongly repressed the IRF1-mediated induction of Trim21. Consistent with this observation, baseline expression of Trim21 was elevated in Irf4(-/-) cells. TRIM21, IRF1, and IRF2 expression was increased in PBMCs from patients with Sjögren's syndrome compared with healthy controls. In contrast, IRF4 and IRF8 expression was not increased in PBMCs from patients. The IFN-γ-mediated induction of Trim21 was completely abolished by inhibiting protein synthesis with cycloheximide, and Trim21 expression could not be induced by IFN-γ in Irf1(-/-) cells, demonstrating that IFN-γ induces Trim21 indirectly via IRF1 and not directly via STAT1 activation. Our data demonstrate that multiple IRFs tightly regulate expression of Trim21 in immune cells, suggesting that a well-controlled expression of the E3 ligase TRIM21 is important for regulation of immune responses.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Ribonucleoproteínas/genética , Animales , Secuencia de Bases , Línea Celular , Secuencia Conservada , Orden Génico , Humanos , Factor 1 Regulador del Interferón/metabolismo , Factor 2 Regulador del Interferón/metabolismo , Interferón-alfa/farmacología , Interferón beta/farmacología , Interferón gamma/farmacología , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Elementos de Respuesta , Factor de Transcripción STAT1/metabolismo , Síndrome de Sjögren/genética , Síndrome de Sjögren/metabolismo
19.
Eur J Immunol ; 42(9): 2274-84, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22949326

RESUMEN

Systemic autoimmune diseases such as systemic lupus erythematosus are type I IFN-driven diseases with exaggerated B-cell responses and autoantibody production. Th17 cells, a T-helper-cell subset with high inflammatory capacity, was initially discovered and characterized in the context of experimental autoimmune encephalomyelitis - an animal model of multiple sclerosis. There is now emerging evidence that Th17 cells, and more generally IL-17 and IL-17-producing cells, may play a role in the pathogenesis of type I IFN-driven systemic autoimmune diseases such as lupus. Here, we review the different studies suggesting a role for IL-17 and IL-17-producing cells in systemic autoimmune diseases, both in humans and in animal models, and we consider the possible mechanisms by which these cells may contribute to disease. We also discuss the hypothesis that type I IFN and IL-17 act in concert to sustain and amplify autoimmune and inflammatory responses, making them a dangerous combination involved in the pathogenesis of systemic autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Interferón Tipo I/inmunología , Interleucina-17/inmunología , Células Th17/inmunología , Animales , Humanos
20.
Arthritis Rheum ; 64(5): 1601-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22127978

RESUMEN

OBJECTIVE: To examine the role of interferon regulatory factor 3 (IRF-3) in the regulation of interleukin-23 (IL-23) production in patients with systemic lupus erythematosus (SLE). METHODS: Bone marrow-derived macrophages were isolated from both wild-type and IRF3(-/-) C57BL/6 mice. These cells were stimulated with the Toll-like receptor 3 (TLR-3) agonist poly(I-C), and IL-23p19 cytokine levels were analyzed by enzyme-linked immunosorbent assay. IRF-3 binding to the IL-23p19 gene promoter region in monocytes from patients with SLE and healthy control subjects was analyzed by chromatin immunoprecipitation (ChIP) assay. Luciferase reporter gene assays were performed to identify key drivers of IL-23p19 promoter activity. TANK-binding kinase 1 (TBK-1) protein levels were determined by Western blotting. RESULTS: ChIP assays demonstrated that IRF-3 was stably bound to the human IL-23p19 promoter in monocytes; this association increased following TLR-3 stimulation. Patients with SLE demonstrated increased levels of IRF-3 bound to the IL-23p19 promoter compared with control subjects, which correlated with enhanced IL-23p19 production in monocytes from patients with SLE. Investigations of the TLR-3-driven responses in monocytes from patients with SLE revealed that TBK-1, which is critical for regulating IRF-3 activity, was hyperactivated in both resting and TLR-3-stimulated cells. CONCLUSION: Our results demonstrate for the first time that patients with SLE display enhanced IL-23p19 expression as a result of hyperactivation of TBK-1, resulting in increased binding of IRF-3 to the promoter. These findings provide novel insights into the molecular pathogenesis of SLE and the potential role for TLR-3 in driving this response.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Subunidad p19 de la Interleucina-23/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/genética , Subunidad p19 de la Interleucina-23/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Poli I-C/farmacología , Análisis por Matrices de Proteínas/métodos , Unión Proteica , Proteínas Serina-Treonina Quinasas/farmacología , Receptor Toll-Like 3/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA