Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Rheumatology (Oxford) ; 63(2): 551-562, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37341646

RESUMO

OBJECTIVES: Platelets and low-density neutrophils (LDNs) are major players in the immunopathogenesis of SLE. Despite evidence showing the importance of platelet-neutrophil complexes (PNCs) in inflammation, little is known about the relationship between LDNs and platelets in SLE. We sought to characterize the role of LDNs and Toll-like receptor 7 (TLR7) in clinical disease. METHODS: Flow cytometry was used to immunophenotype LDNs from SLE patients and controls. The association of LDNs with organ damage was investigated in a cohort of 290 SLE patients. TLR7 mRNA expression was assessed in LDNs and high-density neutrophils (HDNs) using publicly available mRNA sequencing datasets and our own cohort using RT-PCR. The role of TLR7 in platelet binding was evaluated in platelet-HDN mixing studies using TLR7-deficient mice and Klinefelter syndrome patients. RESULTS: SLE patients with active disease have more LDNs, which are heterogeneous and more immature in patients with evidence of kidney dysfunction. LDNs are platelet bound, in contrast to HDNs. LDNs settle in the peripheral blood mononuclear cell (PBMC) layer due to the increased buoyancy and neutrophil degranulation from platelet binding. Mixing studies demonstrated that this PNC formation was dependent on platelet-TLR7 and that the association results in increased NETosis. The neutrophil:platelet ratio is a useful clinical correlate for LDNs, and a higher NPR is associated with past and current flares of LN. CONCLUSIONS: LDNs sediment in the upper PBMC fraction due to PNC formation, which is dependent on the expression of TLR7 in platelets. Collectively, our results reveal a novel TLR7-dependent crosstalk between platelets and neutrophils that may be an important therapeutic opportunity for LN.


Assuntos
Nefrite Lúpica , Neutrófilos , Animais , Humanos , Camundongos , Leucócitos Mononucleares , Nefrite Lúpica/patologia , Neutrófilos/metabolismo , RNA Mensageiro/metabolismo , Receptor 7 Toll-Like/genética
2.
Rheumatology (Oxford) ; 60(8): 3936-3944, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-33369679

RESUMO

OBJECTIVES: To identify and quantify the level of CD34+CD133+CD309+ circulating angiogenic cells (CAC) and explore factors associated with the level of CAC in patients with SLE. METHODS: The peripheral blood mononuclear cells of consecutive SLE patients and demographically matched healthy controls (HC) were extracted and identified, enumerated and compared for CAC levels by multi-colour flow cytometry based on the EULAR Scleroderma Trials and Research (EUSTAR) recommendation. Meta-analyses were performed by combining the current and previous case-control studies, aiming to increase the statistical power to discern the difference in CAC level between SLE patients and HC. Mixed-model meta-regression was conducted to explore potential demographic and clinical factors that were associated with CAC level. RESULTS: A lower level of CAC was found in 29 SLE patients compared with 24 HC [mean (s.d.) 10.76 (13.9) vs 24.58 (25.4) cells/ml, P = 0.015]. Random-effects meta-analyses of the current and six previously published case-control studies involving 401 SLE patients and 228 HC revealed a lower CAC level compared with HC (standardized mean difference = -2.439, P = 0.001). Meta-regression analysis demonstrated that HCQ use was associated with a more discrepant CAC level between both groups (P = 0.01115). CONCLUSION: SLE patients had a significantly lower CD34+CD133+CD309+ CAC level than HC, and HCQ use was associated with a more discrepant CAC level between SLE patients and HC. This study triggers further observational, interventional and mechanistic studies to address the beneficial impact of HCQ on the functionality of CAC in SLE patients.


Assuntos
Células Progenitoras Endoteliais/citologia , Lúpus Eritematoso Sistêmico/sangue , Antígeno AC133/metabolismo , Adulto , Antígenos CD34/metabolismo , Antirreumáticos/uso terapêutico , Estudos de Casos e Controles , Células Progenitoras Endoteliais/metabolismo , Feminino , Citometria de Fluxo , Humanos , Hidroxicloroquina/uso terapêutico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
3.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255483

RESUMO

In eukaryotes, microRNAs (miRNAs) have roles in development, homeostasis, disease and the immune response. Recent work has shown that plant and mammalian miRNAs also mediate cross-kingdom and cross-domain communications. However, these studies remain controversial and are lacking critical mechanistic explanations. Bacteria do not produce miRNAs themselves, and therefore it is unclear how these eukaryotic RNA molecules could function in the bacterial recipient. In this review, we compare and contrast the biogenesis and functions of regulatory RNAs in eukaryotes and bacteria. As a result, we discovered several conserved features and homologous components in these distinct pathways. These findings enabled us to propose novel mechanisms to explain how eukaryotic miRNAs could function in bacteria. Further understanding in this area is necessary to validate the findings of existing studies and could facilitate the use of miRNAs as novel tools for the directed remodelling of the human microbiota.


Assuntos
Bactérias/genética , Eucariotos/genética , MicroRNAs/genética , RNA/genética , Humanos , Microbiota/genética
4.
Cytometry A ; 95(3): 268-278, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30549398

RESUMO

Neutrophil extracellular traps (NETs) are web-like structures composed of decondensed chromatin and antimicrobial proteins that are released into the extracellular space during microbial infections. This active cell death program is known as NETosis. To date, florescence microscopy is the widely accepted method for visualization and quantification of NETs. However, this method is subjective, time consuming and yields low numbers of analyzed polymorphonuclear cells (PMNs) per sample. Increasing interest has emerged on the identification of NETs using flow cytometry techniques. However, flow cytometry analysis of NETs requires particular precautions for sample preparation to obtain reproducible data. Herein, we describe a flow cytometry-based assay for high-throughput detection and quantification of NETosis in mixed cell populations. We used fluorescent-labeled antibodies against cell markers on PMNs together with a combination of nucleic acid stains to measure NETosis in whole blood (WB) and purified PMNs. Using plasma membrane-impermeable DNA-binding dye, SYTOX Orange (SO), we found that cell-appendant DNA of NETting PMNs were positive for SO and DAPI. The combination of optimally diluted antibody and nucleic acid dyes required no washing and yielded low background fluorescence. Significant correlations were found for NETosis from WB and purified PMNs. We then validated the assay by comparing with time-lapse live cell fluorescence microscopy and determined very good intraassay and interassay variances. The assay was then applied to a disease associated with NETosis, systemic lupus erythematosus (SLE). We examined PMA-induced NETosis in peripheral PMNs from SLE patients and controls and in bone marrow PMNs from multiple murine models. In summary, this assay is observer-independent and allows for rapid assessment of a large number of PMNs per sample. Use of this assay does not require sophisticated microscopic equipment like imaging flow cytometers and may be a starting point to analyze extracellular trap formation from immune cells other than PMNs. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Assuntos
Armadilhas Extracelulares/metabolismo , Citometria de Fluxo/métodos , Imunofluorescência/métodos , Microscopia de Fluorescência/métodos , Neutrófilos/metabolismo , Animais , Células da Medula Óssea/metabolismo , DNA/análise , DNA/química , Modelos Animais de Doenças , Armadilhas Extracelulares/química , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Morte Celular Regulada/efeitos dos fármacos , Morte Celular Regulada/genética
5.
Proc Natl Acad Sci U S A ; 112(45): E6195-204, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26512111

RESUMO

Glomerulonephritis is a common and debilitating feature of systemic lupus erythematosus (SLE). The precise immune mechanisms that drive the progression from benign autoimmunity to glomerulonephritis are largely unknown. Previous investigations have shown that a moderate increase of the innate Toll-like receptor 7 (TLR7) is sufficient for the development of nephritis. In these systems normalization of B-cell TLR7 expression or temporal depletion of plasmacytoid dendritic cells (pDCs) slow progression; however, the critical cell that is responsible for driving full immunopathology remains unidentified. In this investigation we have shown that conventional DC expression of TLR7 is essential for severe autoimmunity in the Sle1Tg7 model of SLE. We show that a novel expanding CD11b(+) conventional DC subpopulation dominates the infiltrating renal inflammatory milieu, localizing to the glomeruli. Moreover, exposure of human myeloid DCs to IFN-α or Flu increases TLR7 expression, suggesting they may have a role in self-RNA recognition pathways in clinical disease. To our knowledge, this study is the first to highlight the importance of conventional DC-TLR7 expression for kidney pathogenesis in a murine model of SLE.


Assuntos
Células Dendríticas/metabolismo , Nefrite Lúpica/fisiopatologia , Receptor 7 Toll-Like/metabolismo , Regulação para Cima , Análise de Variância , Animais , Sequência de Bases , Antígeno CD11b/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Humanos , Processamento de Imagem Assistida por Computador , Glomérulos Renais/citologia , Glomérulos Renais/patologia , Nefrite Lúpica/metabolismo , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Estatísticas não Paramétricas
6.
Rheumatology (Oxford) ; 56(suppl_1): i88-i99, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28013204

RESUMO

Mouse models of SLE have been indispensable tools to study disease pathogenesis, to identify genetic susceptibility loci and targets for drug development, and for preclinical testing of novel therapeutics. Recent insights into immunological mechanisms of disease progression have boosted a revival in SLE drug development. Despite promising results in mouse studies, many novel drugs have failed to meet clinical end points. This is probably because of the complexity of the disease, which is driven by polygenic predisposition and diverse environmental factors, resulting in a heterogeneous clinical presentation. Each mouse model recapitulates limited aspects of lupus, especially in terms of the mechanism underlying disease progression. The main mouse models have been fairly successful for the evaluation of broad-acting immunosuppressants. However, the advent of targeted therapeutics calls for a selection of the most appropriate model(s) for testing and, ultimately, identification of patients who will be most likely to respond.


Assuntos
Modelos Animais de Doenças , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Animais , Humanos , Lúpus Eritematoso Sistêmico/genética
7.
Rheumatology (Oxford) ; 56(6): 1025-1030, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28137763

RESUMO

Objective: Siglecs are sialic acid-binding immunoglobulin-like lectins expressed on the surface of immune cells, which participate in the discrimination of self and non-self. We investigated myeloid CD33-related Siglec expression in a cohort of patients with SLE. Methods: Cell surface expression of Siglec-5/14, Siglec-9 and Siglec-10 on peripheral myeloid subsets were analysed from 39 SLE patients using flow cytometry. Genotyping of the Siglec-5/14 locus was also performed. Clinical markers of SLE disease activity, including SLEDAI, serum complement concentrations and serum autoantibodies, were assessed and correlated with Siglec levels. Results: Siglec-14 expression on SLE monocytes (median = 518, interquartile range: 411) was significantly higher when compared with healthy controls (median = 427, interquartile range: 289.3; P < 0.05) and correlated positively with SLEDAI scoring and anti-Sm and anti-SSB autoantibodies (P < 0.05). A negative correlation was determined with patient serum C3 concentrations (P < 0.005). Genotyping of the Siglec-5/14 locus revealed a high frequency of the Siglec-14 null allele across both groups, reflecting the incidence in Asian populations. Conclusion: Our data suggest that the Siglec immunomodulatory molecules, in particular Siglec-14 expression on monocytes, may play an important role in the inflammatory events of SLE. No bias was found with regard to SIGLEC14 genotype in our patient group compared with healthy controls. Larger comparisons of mixed ethnicity might, however, reveal an important role for Siglecs in the pathogenesis of autoimmune disease.


Assuntos
Lectinas/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Receptores de Superfície Celular/metabolismo , Adulto , Anti-Inflamatórios/uso terapêutico , Estudos de Casos e Controles , Feminino , Genótipo , Heterozigoto , Humanos , Lectinas/genética , Leucócitos Mononucleares/metabolismo , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/etnologia , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético , Prednisolona/uso terapêutico , Receptores de Superfície Celular/genética , Regulação para Cima/fisiologia , Adulto Jovem
8.
Rheumatology (Oxford) ; 56(suppl_1): i55-i66, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28375453

RESUMO

SLE is a chronic autoimmune disease caused by perturbations of the immune system. The clinical presentation is heterogeneous, largely because of the multiple genetic and environmental factors that contribute to disease initiation and progression. Over the last 60 years, there have been a number of significant leaps in our understanding of the immunological mechanisms driving disease processes. We now know that multiple leucocyte subsets, together with inflammatory cytokines, chemokines and regulatory mediators that are normally involved in host protection from invading pathogens, contribute to the inflammatory events leading to tissue destruction and organ failure. In this broad overview, we discuss the main pathways involved in SLE and highlight new findings. We describe the immunological changes that characterize this form of autoimmunity. The major leucocytes that are essential for disease progression are discussed, together with key mediators that propagate the immune response and drive the inflammatory response in SLE.


Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Citocinas/imunologia , Meio Ambiente , Inflamação/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Autoimunidade/genética , Predisposição Genética para Doença , Humanos , Inflamação/genética , Lúpus Eritematoso Sistêmico/genética , Tolerância a Antígenos Próprios/genética , Tolerância a Antígenos Próprios/imunologia
9.
Int Immunol ; 28(5): 223-32, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26567289

RESUMO

The toll-like receptors (TLRs) are important innate receptors recognizing potentially pathogenic material. However, they also play a significant role in the development of Alzheimer's disease, cancer, autoimmunity and the susceptibility to viral infections. Macrophages are essential for an effective immune response to foreign material and the resolution of inflammation. In these studies, we examined the impact of different TLR ligands on macrophage cell function. We demonstrate that stimulation of all TLRs tested increases the phagocytosis of apoptotic cells by macrophages. TLR7 and TLR9 ligation decreased the levels of the surface co-expression molecules CD86 and MHCII, which was associated with a concomitant reduction in antigen presentation and proliferation of T cells. This down-regulation in macrophage function was not due to an increase in cell death. In fact, exposure to TLR7 or TLR9 ligands promoted cell viability for up to 9 days, in contrast to TLR3 or TLR4. Additionally, macrophages exposed to TLR7/TLR9 ligands had a significantly lower ratio of Il-12/Il-10 mRNA expression compared with those treated with the TLR4 ligand, LPS. Taken together, these data demonstrate that TLR7/TLR9 ligands push the macrophage into a phagocytic long-lived cell, with a decreased capacity of antigen presentation and reminiscent of the M2 polarized state.


Assuntos
Apresentação de Antígeno , Macrófagos/imunologia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/imunologia , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/imunologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Ligantes , Lipopolissacarídeos/toxicidade , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Fagocitose/efeitos dos fármacos , Fagocitose/genética , Receptor 7 Toll-Like/genética , Receptor Toll-Like 9/genética
10.
J Immunol ; 194(11): 5174-86, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25917094

RESUMO

Human dendritic cells (DCs) regulate the balance between immunity and tolerance through selective activation by environmental and pathogen-derived triggers. To characterize the rapid changes that occur during this process, we analyzed the underlying metabolic activity across a spectrum of functional DC activation states, from immunogenic to tolerogenic. We found that in contrast to the pronounced proinflammatory program of mature DCs, tolerogenic DCs displayed a markedly augmented catabolic pathway, related to oxidative phosphorylation, fatty acid metabolism, and glycolysis. Functionally, tolerogenic DCs demonstrated the highest mitochondrial oxidative activity, production of reactive oxygen species, superoxide, and increased spare respiratory capacity. Furthermore, assembled, electron transport chain complexes were significantly more abundant in tolerogenic DCs. At the level of glycolysis, tolerogenic and mature DCs showed similar glycolytic rates, but glycolytic capacity and reserve were more pronounced in tolerogenic DCs. The enhanced glycolytic reserve and respiratory capacity observed in these DCs were reflected in a higher metabolic plasticity to maintain intracellular ATP content. Interestingly, tolerogenic and mature DCs manifested substantially different expression of proteins involved in the fatty acid oxidation (FAO) pathway, and FAO activity was significantly higher in tolerogenic DCs. Inhibition of FAO prevented the function of tolerogenic DCs and partially restored T cell stimulatory capacity, demonstrating their dependence on this pathway. Overall, tolerogenic DCs show metabolic signatures of increased oxidative phosphorylation programing, a shift in redox state, and high plasticity for metabolic adaptation. These observations point to a mechanism for rapid genome-wide reprograming by modulation of underlying cellular metabolism during DC differentiation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Tolerância Imunológica/imunologia , Mitocôndrias/metabolismo , Consumo de Oxigênio , 3-Hidroxiacil-CoA Desidrogenases/antagonistas & inibidores , 3-Hidroxiacil-CoA Desidrogenases/genética , Acetil-CoA C-Aciltransferase/antagonistas & inibidores , Acetil-CoA C-Aciltransferase/genética , Isomerases de Ligação Dupla Carbono-Carbono/antagonistas & inibidores , Isomerases de Ligação Dupla Carbono-Carbono/genética , Diferenciação Celular , Células Cultivadas , Complexo de Proteínas da Cadeia de Transporte de Elétrons/biossíntese , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Enoil-CoA Hidratase/antagonistas & inibidores , Enoil-CoA Hidratase/genética , Ácidos Graxos/metabolismo , Glicólise , Humanos , Leucócitos Mononucleares/imunologia , Oxirredução , Fosforilação Oxidativa , Racemases e Epimerases/antagonistas & inibidores , Racemases e Epimerases/genética , Superóxidos/metabolismo , Linfócitos T/imunologia
11.
J Immunol ; 191(12): 6128-35, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24218451

RESUMO

The three-prime repair exonuclease 1 (TREX1) is the most abundant exonuclease in mammalian cells. Mutations in Trex1 gene are being linked to the development of Aicardi-Goutières syndrome, an inflammatory disease of the brain, and systemic lupus erythematosus. In clinical cases and in a Trex1-deficient murine model, chronic production of type I IFN plays a pathogenic role. In this study, we demonstrate that Trex1(-/-) mice present inflammatory signatures in many different organs, including the brain. Trex1 is highly induced in macrophages in response to proinflammatory stimuli, including TLR7 and TLR9 ligands. Our findings show that, in the absence of Trex1, macrophages displayed an exacerbated proinflammatory response. More specifically, following proinflammatory stimulation, Trex1(-/-) macrophages exhibited an increased TNF-α and IFN-α production, higher levels of CD86, and increased Ag presentation to CD4(+) T cells, as well as an impaired apoptotic T cell clearance. These results evidence an unrevealed function of the Trex1 as a negative regulator of macrophage inflammatory activation and demonstrate that macrophages play a major role in diseases associated with Trex1 mutations, which contributes to the understanding of inflammatory signature in these diseases.


Assuntos
Exodesoxirribonucleases/fisiologia , Inflamação/imunologia , Ativação de Macrófagos/fisiologia , Fosfoproteínas/fisiologia , Animais , Apresentação de Antígeno , Apoptose , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Química Encefálica , Exodesoxirribonucleases/deficiência , Exodesoxirribonucleases/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/metabolismo , Interferon-alfa/biossíntese , Interferon-alfa/genética , Células Jurkat , Células L , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fagocitose , Fosfoproteínas/deficiência , Fosfoproteínas/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T/imunologia , Linfócitos T/patologia , Receptor Toll-Like 9/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
12.
J Immunol ; 191(8): 4375-82, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24048896

RESUMO

TLRs play a pivotal role in the recognition of bacteria and viruses. Members of the family recognize specific pathogen sequences to trigger both MyD88 and TRIF-dependent pathways to stimulate a plethora of cells. Aberrant activation of these pathways is known to play a critical role in the development of autoimmunity and cancer. However, how these pathways are entirely regulated is not fully understood. In these studies, we have identified Annexin-A1 (ANXA1) as a novel regulator of TLR-induced IFN-ß and CXCL10 production. We demonstrate that in the absence of ANXA1, mice produce significantly less IFN-ß and CXCL10, and macrophages and plasmacytoid dendritic cells have a deficiency in activation following polyinosinic:polycytidylic acid administration in vivo. Furthermore, a deficiency in activation is observed in macrophages after LPS and polyinosinic:polycytidylic acid in vitro. In keeping with these findings, overexpression of ANXA1 resulted in enhanced IFN-ß and IFN-stimulated responsive element promoter activity, whereas silencing of ANXA1 impaired TLR3- and TLR4-induced IFN-ß and IFN-stimulated responsive element activation. In addition, we show that the C terminus of ANXA1 directly associates with TANK-binding kinase 1 to regulate IFN regulatory factor 3 translocation and phosphorylation. Our findings demonstrate that ANXA1 plays an important role in TLR activation, leading to an augmentation in the type 1 IFN antiviral cytokine response.


Assuntos
Anexina A1/metabolismo , Interferon beta/biossíntese , Proteínas Serina-Treonina Quinases/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Anexina A1/biossíntese , Anexina A1/genética , Linhagem Celular , Quimiocina CXCL10/biossíntese , Células Dendríticas/metabolismo , Ativação Enzimática , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/metabolismo , Lipopolissacarídeos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosforilação , Poli I-C/farmacologia , Transdução de Sinais/imunologia
13.
J Immunol ; 189(12): 5786-96, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23150717

RESUMO

Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease characterized by the production of antinuclear autoantibodies. Antinuclear autoantibody development is recognized as one of the initial stages of disease that often results in systemic inflammation, kidney disease, and death. The etiology is complex, but it is clear that innate pathways may play an important role in disease progression. Recent data have highlighted an important role for the TLR family, particularly TLR7, in both human disease and murine models. In this study, we have presented a low copy conditional TLR7 transgenic (Tg7) mouse strain that does not develop spontaneous autoimmunity. When we combine Tg7 with the Sle1 lupus susceptibility locus, the mice develop severe disease. Using the CD19(Cre) recombinase system, we normalized expression of TLR7 solely within the B cells. Using this method we demonstrated that overexpression of TLR7 within the B cell compartment reduces the marginal zone B cell compartment and increases B and T cell activation but not T follicular helper cell development. Moreover, this enhanced B cell TLR7 expression permits the specific development of Abs to RNA/protein complexes and exacerbates SLE disease.


Assuntos
Autoanticorpos/biossíntese , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/patologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico/imunologia , Glicoproteínas de Membrana/genética , Receptor 7 Toll-Like/genética , Animais , Autoanticorpos/efeitos adversos , Subpopulações de Linfócitos B/metabolismo , Progressão da Doença , Epistasia Genética/imunologia , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Receptor 7 Toll-Like/biossíntese , Receptor 7 Toll-Like/fisiologia , Transgenes/imunologia
15.
Front Immunol ; 13: 807050, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154124

RESUMO

Cytokine release syndrome (CRS) is a phenomenon of immune hyperactivation described in the setting of immunotherapy. Unlike other immune-related adverse events, CRS triggered by immune checkpoint inhibitors (ICIs) is not well described. The clinical characteristics and course of 25 patients with ICI-induced CRS from 2 tertiary hospitals were abstracted retrospectively from the medical records and analyzed. CRS events were confirmed by 2 independent reviewers and graded using the Lee et al. scale. The median duration of CRS was 15.0 days (Q1; Q3 6.3; 29.8) and 10 (40.0%) had multiple episodes of CRS flares. Comparing the clinical factors and biomarkers in Grades 1-2 and 3-5 CRS, we found that patients with Grades 3-5 CRS had following: (i) had longer time to fever onset [25.0 days (Q1; Q3 13.0; 136.5) vs. 3.0 days (Q1; Q3 0.0; 18.0), p=0.027]; (ii) more cardiovascular (p=0.002), neurologic (p=0.001), pulmonary (p=0.044) and rheumatic (p=0.037) involvement; (iii) lower platelet count (p=0.041) and higher urea (p=0.041) at presentation compared to patients with Grades 1-2 CRS. 7 patients (28.0%) with Grades 1-2 CRS were rechallenged using ICIs without event. 9 patients (36.0%) were treated with pulse methylprednisolone and 6 patients (24.0%) were treated with tocilizumab. Despite this, 3 patients (50%) who received tocilizumab had fatal (Grade 5) outcomes from ICI-induced CRS. Longer time to fever onset, lower platelet count and higher urea at presentation were associated with Grade 3-5 CRS. These parameters may be used to predict which patients are likely to develop severe CRS.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Síndrome da Liberação de Citocina/induzido quimicamente , Síndrome da Liberação de Citocina/tratamento farmacológico , Inibidores de Checkpoint Imunológico/efeitos adversos , Imunoterapia/efeitos adversos , Metilprednisolona/administração & dosagem , Neoplasias/terapia , Índice de Gravidade de Doença , Idoso , Biomarcadores/sangue , Síndrome da Liberação de Citocina/sangue , Evolução Fatal , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pulsoterapia/métodos , Estudos Retrospectivos , Centros de Atenção Terciária , Resultado do Tratamento
16.
Arthritis Rheum ; 62(11): 3436-46, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20722038

RESUMO

OBJECTIVE: CXCR4 is a chemokine with multiple effects on the immune system. In murine lupus models, we demonstrated that monocytes, neutrophils, and B cells overexpressed CXCR4 and that its ligand, CXCL12, was up-regulated in diseased kidneys. We undertook this study to determine whether CXCR4 expression was increased in peripheral blood leukocytes from patients with systemic lupus erythematosus (SLE) and whether CXCL12 expression was increased in kidneys from patients with SLE. METHODS: Peripheral blood leukocytes from 31 SLE patients, 8 normal controls, and 9 patients with rheumatoid arthritis were prospectively analyzed by flow cytometry for CXCR4 expression. Biopsy samples (n = 14) from patients with lupus nephritis (LN) were immunostained with anti-CXCL12 antibody. RESULTS: CD19+ B cells and CD4+ T cells from SLE patients displayed a >2-fold increase (P = 0.0001) and >3-fold increase (P < 0.0001), respectively, in median CXCR4 expression compared with that in controls (n = 7-8). Moreover, CXCR4 expression on B cells was 1.61-fold higher in patients with SLE Disease Activity Index (SLEDAI) scores >10 (n = 8) than in patients with SLEDAI scores ≤10 (n = 16) (P = 0.0008), 1.71-fold higher in patients with class IV LN (n = 5) than in patients with other classes of LN (n = 7) (P = 0.02), and 1.40-fold higher in patients with active neuropsychiatric SLE (NPSLE) (n = 6) than in patients with inactive NPSLE (n = 18) (P = 0.01). CXCL12 was significantly up-regulated in the tubules and glomeruli of kidneys in patients with LN (n = 14), with the percentage of positive cells correlating positively with the severity of LN. CONCLUSION: CXCR4 appears to be up-regulated in multiple leukocyte subsets in SLE patients. The heightened expression of CXCR4 on B cells in active NPSLE and of CXCL12 in nephritic kidneys suggests that the CXCR4/CXCL12 axis might be a potential therapeutic target for SLE patients with kidney and/or central nervous system involvement.


Assuntos
Quimiocina CXCL12/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Receptores CXCR4/metabolismo , Adolescente , Adulto , Idoso , Quimiocina CXCL12/imunologia , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Leucócitos/imunologia , Leucócitos/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/metabolismo , Masculino , Pessoa de Meia-Idade , Receptores CXCR4/imunologia , Regulação para Cima/imunologia
17.
J Immunol ; 182(5): 2700-7, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234164

RESUMO

Preterm birth occurs at a rate of 12.7% in the U.S. and is the primary cause of fetal morbidity in the first year of life as well as the cause of later health problems. Elucidation of mechanisms controlling cervical remodeling is critical for development of therapies to reduce the incidence of prematurity. The cervical extracellular matrix must be disorganized during labor to allow birth, followed by a rapid repair postpartum. Leukocytes infiltrate the cervix before and after birth and are proposed to regulate matrix remodeling during cervical ripening via release of proteolytic enzymes. In the current study, flow cytometry and cell sorting were used to determine the role of immune cells in cervical matrix remodeling before, during, and after parturition. Markers of myeloid cell differentiation and activation were assessed to define phenotype and function. Tissue monocytes and eosinophils increased in the cervix before birth in a progesterone-regulated fashion, whereas macrophage numbers were unchanged. Neutrophils increased in the postpartum period. Increased mRNA expression of Csfr1 and markers of alternatively activated M2 macrophages during labor or shortly postpartum suggest a function of M2 macrophages in postpartum tissue repair. Changes in cervical myeloid cell numbers are not reflected in the peripheral blood. These data along with our previous studies suggest that myeloid-derived cells do not orchestrate processes required for initiation of cervical ripening before birth. Additionally, macrophages with diverse phenotypes (M1 and M2) are present in the cervix and are most likely involved in the postpartum repair of tissue.


Assuntos
Colo do Útero/citologia , Colo do Útero/imunologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Parto/imunologia , Parto/metabolismo , Animais , Animais Recém-Nascidos , Movimento Celular/imunologia , Colo do Útero/metabolismo , Eosinófilos/imunologia , Eosinófilos/metabolismo , Feminino , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Contagem de Leucócitos , Ativação de Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Células Mieloides/citologia , Gravidez , Fatores de Tempo , Cicatrização/imunologia
18.
J Immunol ; 182(7): 4448-58, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299746

RESUMO

Among various surface molecules screened, CXCR4 was significantly up-regulated on monocytes, neutrophils, B cell subsets, and plasma cells in multiple murine models of lupus with active nephritis, including B6.Sle1Yaa, BXSB, and MRL.lpr. TLR-mediated signaling and inflammatory cytokines accounted in part for this increase. Increased CXCR4 expression was associated with functional consequences, including increased migration and enhanced B cell survival. Simultaneously, the ligand for CXCR4, CXCL12, was significantly up-regulated in the nephritic kidneys. Treatment with a peptide antagonist of CXCR4 prolonged survival and reduced serum autoantibodies, splenomegaly, intrarenal leukocyte trafficking, and end organ disease in a murine model of lupus. These findings underscore the pathogenic role of CXCR4/CXCL12 in lymphoproliferative lupus and lupus nephritis and highlight this axis as a promising therapeutic target in this disease.


Assuntos
Quimiocina CXCL12/biossíntese , Leucócitos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Receptores CXCR4/biossíntese , Animais , Quimiocina CXCL12/imunologia , Quimiotaxia de Leucócito/imunologia , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Leucócitos/metabolismo , Lúpus Eritematoso Sistêmico/complicações , Nefrite Lúpica/etiologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Masculino , Camundongos , Receptores CXCR4/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Regulação para Cima
19.
J Immunol ; 183(10): 6831-8, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864599

RESUMO

Increased Type I IFNs or IFN-I have been associated with human systemic lupus erythematosus. Interestingly augmenting or negating IFN-I activity in murine lupus not only modulates systemic autoimmunity, but also impacts lupus nephritis, suggesting that IFN-I may be acting at the level of the end-organ. We find resident renal cells to be a dominant source of IFN-I in an experimental model of autoantibody-induced nephritis. In this model, augmenting IFN-I amplified antibody-triggered nephritis, whereas ablating IFN-I activity ameliorated disease. One mechanism through which increased IFN-I drives immune-mediated nephritis might be operative through increased recruitment of inflammatory monocytes and neutrophils, though this hypothesis needs further validation. Collectively, these studies indicate that an important contribution of IFN-I toward the disease pathology seen in systemic autoimmunity may be exercised at the level of the end-organ.


Assuntos
Citocinas/imunologia , Glomerulonefrite/imunologia , Interferon Tipo I/imunologia , Falência Renal Crônica/imunologia , Receptor de Interferon alfa e beta/imunologia , Animais , Antivirais/imunologia , Antivirais/farmacologia , Autoanticorpos/sangue , Autoanticorpos/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Glomerulonefrite/metabolismo , Humanos , Interferon Tipo I/farmacologia , Rim/efeitos dos fármacos , Rim/imunologia , Rim/patologia , Falência Renal Crônica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Proteínas Recombinantes
20.
Curr Protoc ; 1(11): e272, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34748281

RESUMO

Systemic Lupus Erythematosus (SLE) is a complex and heterogenous autoimmune disease, where genetics, immunology, and environmental factors all play a role. Murine models have contributed critical information on mechanisms of disease and prospective therapeutics. The key features that have been used to study the disease include the development of anti-nuclear autoantibodies (ANAs), splenomegaly, and kidney disease. The loss of tolerance and subsequent autoimmune features, and the progression to severe disease, are all dependent on immune dysregulation. In this article, we will describe the methods used to evaluate the underlying immunological features of the disease, as a more sensitive strategy to understand the disease itself and the mechanisms of potential novel therapeutics. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: End study protocols for tissue harvesting Basic Protocol 2: End study protocols for tissue processing Basic Protocol 3: Immunophenotyping using flow cytometry protocols Support Protocol: Tissue processing for cold storage Basic Protocol 4: Additional tissue processing for later analyses Basic Protocol 5: Analysis of serum auto-antibodies by ELISAs (ANAs, snRNP, and dsDNA).


Assuntos
Doenças Autoimunes , Lúpus Eritematoso Discoide , Lúpus Eritematoso Sistêmico , Animais , Autoanticorpos , Modelos Animais de Doenças , Camundongos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa