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

Tipo de documento
Intervalo de ano de publicação
1.
EMBO Rep ; 24(1): e54944, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36341538

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Melanoma , Humanos , Linhagem Celular Tumoral , Proteína 28 com Motivo Tripartido/genética , Melanoma/genética
2.
Ann Rheum Dis ; 81(8): 1151-1161, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35470161

RESUMO

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.


Assuntos
Bloqueio Atrioventricular , Auxilinas , Animais , Anticorpos Antinucleares , Bloqueio Atrioventricular/genética , Autoanticorpos , Coração Fetal , Estudo de Associação Genômica Ampla , Bloqueio Cardíaco/congênito , Lúpus Eritematoso Sistêmico/congênito , Camundongos
3.
J Biol Chem ; 294(30): 11404-11419, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31160341

RESUMO

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.


Assuntos
Lisina/metabolismo , Ribonucleoproteínas/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Estrutura Molecular , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ribonucleoproteínas/química , Alinhamento de Sequência , Especificidade por Substrato , Enzimas de Conjugação de Ubiquitina/química
4.
Immunology ; 159(3): 335-343, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31755557

RESUMO

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.


Assuntos
Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Ribonucleoproteínas/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Interações Hospedeiro-Patógeno , Lipopeptídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium bovis/imunologia , Mycobacterium bovis/patogenicidade , Fenótipo , Ribonucleoproteínas/deficiência , Ribonucleoproteínas/genética , Transdução de Sinais , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética
5.
Eur J Immunol ; 49(2): 313-322, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30307034

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Metabolismo Energético/imunologia , MicroRNAs/imunologia , Síndrome de Sjogren/imunologia , Linfócitos T CD4-Positivos/patologia , Metabolismo Energético/efeitos dos fármacos , Feminino , Humanos , Interferon-alfa/imunologia , Interferon-alfa/farmacologia , Interferon beta/imunologia , Interferon beta/farmacologia , Masculino , Síndrome de Sjogren/patologia
6.
Gac Med Mex ; 156(4): 334-338, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32831328

RESUMO

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.


Assuntos
Infecções por Coronavirus/epidemiologia , Atenção à Saúde/organização & administração , Pneumonia Viral/epidemiologia , Saúde Pública , Betacoronavirus/isolamento & purificação , COVID-19 , Colômbia/epidemiologia , Humanos , Pandemias , Qualidade de Vida , SARS-CoV-2
7.
J Immunol ; 196(1): 91-6, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26590315

RESUMO

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.


Assuntos
Fator Ativador de Células B/imunologia , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 2 de Interferon/imunologia , Fatores Reguladores de Interferon/imunologia , Interferon beta/imunologia , Autoanticorpos/imunologia , Fator Ativador de Células B/sangue , Fator Ativador de Células B/genética , Linfócitos B/imunologia , Sequência de Bases , Sítios de Ligação/genética , Células Cultivadas , Feminino , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Dados de Sequência Molecular , Monócitos/imunologia , Esclerose Múltipla/imunologia , Neutrófilos/imunologia , Síndrome de Sjogren/imunologia , Transcrição Gênica/genética
8.
Exp Dermatol ; 26(8): 713-720, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27943421

RESUMO

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.


Assuntos
Aminoquinolinas/efeitos adversos , Antineoplásicos/efeitos adversos , Psoríase/induzido quimicamente , Ribonucleoproteínas/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Imiquimode , Camundongos Knockout , Psoríase/metabolismo , Ribonucleoproteínas/genética
9.
Biochem Biophys Res Commun ; 473(4): 789-794, 2016 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-27003259

RESUMO

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.


Assuntos
Aminoquinolinas/farmacologia , Cálcio/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Queratinócitos/fisiologia , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo , Animais , Antineoplásicos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Imiquimode , Queratinócitos/efeitos dos fármacos , Camundongos
10.
J Immunol ; 191(7): 3753-63, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23975864

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Fatores Reguladores de Interferon/metabolismo , Ribonucleoproteínas/genética , Animais , Sequência de Bases , Linhagem Celular , Sequência Conservada , Ordem dos Genes , Humanos , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 2 de Interferon/metabolismo , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Interferon gama/farmacologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Elementos de Resposta , Fator de Transcrição STAT1/metabolismo , Síndrome de Sjogren/genética , Síndrome de Sjogren/metabolismo
11.
Adv Sci (Weinh) ; 11(28): e2401772, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38767114

RESUMO

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.


Assuntos
Androgênios , Modelos Animais de Doenças , Síndrome do Ovário Policístico , Animais , Síndrome do Ovário Policístico/imunologia , Síndrome do Ovário Policístico/metabolismo , Feminino , Camundongos , Androgênios/metabolismo , Camundongos Endogâmicos C57BL , Hiperandrogenismo/imunologia , Hiperandrogenismo/metabolismo
12.
Eur J Immunol ; 42(9): 2274-84, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22949326

RESUMO

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.


Assuntos
Doenças Autoimunes/imunologia , Interferon Tipo I/imunologia , Interleucina-17/imunologia , Células Th17/imunologia , Animais , Humanos
14.
Arthritis Rheum ; 64(5): 1601-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22127978

RESUMO

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.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Subunidade p19 da Interleucina-23/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/genética , Subunidade p19 da Interleucina-23/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Poli I-C/farmacologia , Análise Serial de Proteínas/métodos , Ligação Proteica , Proteínas Serina-Treonina Quinases/farmacologia , Receptor 3 Toll-Like/imunologia
15.
bioRxiv ; 2023 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-37986968

RESUMO

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.

16.
J Biol Chem ; 286(42): 36478-91, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21862588

RESUMO

Ro52 (TRIM21) is an E3 ligase of the tripartite motif family that negatively regulates proinflammatory cytokine production by ubiquitinating transcription factors of the interferon regulatory factor family. Autoantibodies to Ro52 are present in patients with lupus and Sjögren's syndrome, but it is not known if these autoantibodies affect the function of Ro52. To address this question, the requirements for Ro52 E3 ligase activity were first analyzed in detail. Scanning a panel of E2 ubiquitin-conjugating enzymes, we found that UBE2D1-4 and UBE2E1-2 supported the E3 ligase activity of Ro52 and that the E3 ligase activity of Ro52 was dependent on its RING domain. We also found that the N-terminal extensions in the class III E2 enzymes affected their interaction with Ro52. Although the N-terminal extension in UBE2E3 made this E2 enzyme unable to function together with Ro52, the N-terminal extensions in UBE2E1 and UBE2E2 allowed for a functional interaction with Ro52. Anti-Ro52-positive patient sera and affinity-purified anti-RING domain autoantibodies inhibited the E3 activity of Ro52 in ubiquitination assays. Using NMR, limited proteolysis, ELISA, and Ro52 mutants, we mapped the interactions between Ro52, UBE2E1, and anti-Ro52 autoantibodies. We found that anti-Ro52 autoantibodies inhibited the E3 ligase activity of Ro52 by sterically blocking the E2/E3 interaction between Ro52 and UBE2E1. Our data suggest that anti-Ro52 autoantibodies binding the RING domain of Ro52 may be actively involved in the pathogenesis of rheumatic autoimmune disease by inhibiting Ro52-mediated ubiquitination.


Assuntos
Autoanticorpos/imunologia , Ribonucleoproteínas/antagonistas & inibidores , Ribonucleoproteínas/imunologia , Síndrome de Sjogren/imunologia , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação/imunologia , Autoanticorpos/farmacologia , Linhagem Celular , Humanos , Estrutura Terciária de Proteína , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Síndrome de Sjogren/enzimologia , Síndrome de Sjogren/genética , Enzimas de Conjugação de Ubiquitina/antagonistas & inibidores , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos dos fármacos
17.
Biomolecules ; 12(6)2022 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-35740904

RESUMO

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.


Assuntos
Proteína HMGB1 , Ativação de Macrófagos , Transcriptoma , Animais , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos
18.
Nat Commun ; 13(1): 4983, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36127330

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental , Infecções por Escherichia coli , Infecções Urinárias , Animais , Peptídeos Antimicrobianos , Escherichia coli/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Estradiol/metabolismo , Glucose/metabolismo , Humanos , Insulina/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteína A7 Ligante de Cálcio S100/metabolismo , Bexiga Urinária/metabolismo
19.
Mult Scler ; 16(9): 1056-65, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20354066

RESUMO

Definition of dysregulated immune components in multiple sclerosis may help in the identification of new therapeutic targets. Deviation of the interleukin 18 receptor 1 (IL18R1) is of particular interest since the receptor is critical for experimental neuroinflammation. The objective of this study was to determine whether expression of IL18R1 varies between multiple sclerosis patients and controls, and to test genetic association of IL18R1 with multiple sclerosis. We used quantitative real-time PCR to assess mRNA levels of IL18R1 in cerebrospinal fluid and peripheral blood mononuclear cells of 191 patients with multiple sclerosis, 61 patients with clinically isolated syndrome and 168 controls having other neurological disorders. Association was tested in 2153 patients with multiple sclerosis and 1733 controls using 13 tagging single nucleotide polymorphisms within the IL18R1 gene. We found that patients with multiple sclerosis had increased IL18R1 mRNA expression in both cerebrospinal fluid cells (p < 0.05) and peripheral blood mononuclear cells (p < 0.05) compared with controls. Patients with clinically isolated syndrome had elevated levels compared with controls in cerebrospinal fluid cells (p < 0.001) but not in peripheral blood mononuclear cells. The gene was not associated to multiple sclerosis. We conclude that the increased expression of IL18R1 may contribute pathogenically to disease and is therefore a potential therapeutic target. The absence of a genetic association in the IL18R1 gene itself suggests regulation from other parts of the genome, or as part of the inflammatory cascade in multiple sclerosis without a prime genetic cause.


Assuntos
Subunidade alfa de Receptor de Interleucina-18/genética , Esclerose Múltipla Crônica Progressiva/genética , Esclerose Múltipla Recidivante-Remitente/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Estudos de Associação Genética , Haplótipos , Humanos , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Crônica Progressiva/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , RNA Mensageiro/sangue , RNA Mensageiro/líquido cefalorraquidiano , Suécia , Regulação para Cima , Adulto Jovem
20.
RMD Open ; 6(1)2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31958275

RESUMO

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.


Assuntos
Anticorpos Antinucleares/imunologia , Bloqueio Cardíaco/congênito , Interferon Tipo I/imunologia , Adulto , Autoanticorpos/sangue , Autoanticorpos/imunologia , Estudos de Casos e Controles , Ecocardiografia Doppler , Feminino , Bloqueio Cardíaco/sangue , Bloqueio Cardíaco/embriologia , Bloqueio Cardíaco/imunologia , Humanos , Recém-Nascido , Interferon Tipo I/sangue , Masculino , Troca Materno-Fetal/imunologia , Gravidez , Complicações na Gravidez/imunologia , Doenças Reumáticas/imunologia , Suécia , Transcriptoma , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa