Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1345515, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38469292

RESUMO

Background: Chronic Lymphocytic Leukemia (CLL) is characterized by the expansion of CD19+ CD5+ B cells but its origin remains debated. Mutated CLL may originate from post-germinal center B cells and unmutated CLL from CD5+ mature B cell precursors. Irrespective of precursor types, events initiating CLL remain unknown. The cytokines BAFF and APRIL each play a significant role in CLL cell survival and accumulation, but their involvement in disease initiation remains unclear. Methods: We generated novel CLL models lacking BAFF or APRIL. In vivo experiments were conducted to explore the impact of BAFF or APRIL loss on leukemia initiation, progression, and dissemination. Additionally, RNA-seq and quantitative real-time PCR were performed to unveil the transcriptomic signature influenced by BAFF in CLL. The direct role of BAFF in controlling the expression of tumor-promoting genes was further assessed in patient-derived primary CLL cells ex-vivo. Results: Our findings demonstrate a crucial role for BAFF, but not APRIL, in the initiation and dissemination of CLL cells. In the absence of BAFF or its receptor BAFF-R, the TCL1 transgene only increases CLL cell numbers in the peritoneal cavity, without dissemination into the periphery. While BAFF binding to BAFF-R is dispensable for peritoneal CLL cell survival, it is necessary to activate a tumor-promoting gene program, potentially linked to CLL initiation and progression. This direct role of BAFF in controlling the expression of tumor-promoting genes was confirmed in patient-derived primary CLL cells ex-vivo. Conclusions: Our study, involving both mouse and human CLL cells, suggests that BAFF might initiate CLL through mechanisms independent of cell survival. Combining current CLL therapies with BAFF inhibition could offer a dual benefit by reducing peripheral tumor burden and suppressing transformed CLL cell output.


Assuntos
Leucemia Linfocítica Crônica de Células B , Animais , Humanos , Camundongos , Linfócitos B/metabolismo , Sobrevivência Celular/genética , Leucemia Linfocítica Crônica de Células B/patologia
3.
Arterioscler Thromb Vasc Biol ; 43(4): 522-536, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36794587

RESUMO

BACKGROUND: CD4+ (cluster of differentation) and CD8+ T cells are increased in the ocular fluids of patients with neovascular retinopathy, yet their role in the disease process is unknown. METHODS: We describe how CD8+ T cells migrate into the retina and contribute to pathological angiogenesis by releasing cytokines and cytotoxic factors. RESULTS: In oxygen-induced retinopathy, flow cytometry revealed the numbers of CD4+ and CD8+ T cells were increased in blood, lymphoid organs, and retina throughout the development of neovascular retinopathy. Interestingly, the depletion of CD8+ T cells but not CD4+ T cells reduced retinal neovascularization and vascular leakage. Using reporter mice expressing gfp (green fluorescence protein) in CD8+ T cells, these cells were localized near neovascular tufts in the retina, confirming that CD8+ T cells contribute to the disease. Furthermore, the adoptive transfer of CD8+ T cells deficient in TNF (tumor necrosis factor), IFNγ (interferon gamma), Prf (perforin), or GzmA/B (granzymes A/B) into immunocompetent Rag1-/- mice revealed that CD8+ T cells mediate retinal vascular disease via these factors, with TNF influencing all aspects of vascular pathology. The pathway by which CD8+ T cells migrate into the retina was identified as CXCR3 (C-X-C motif chemokine receptor 3) with the CXCR3 blockade reducing the number of CD8+ T cells within the retina and retinal vascular disease. CONCLUSIONS: We discovered that CXCR3 is central to the migration of CD8+ T cells into the retina as the CXCR3 blockade reduced the number of CD8+ T cells within the retina and vasculopathy. This research identified an unappreciated role for CD8+ T cells in retinal inflammation and vascular disease. Reducing CD8+ T cells via their inflammatory and recruitment pathways is a potential treatment for neovascular retinopathies.


Assuntos
Doenças Retinianas , Doenças Vasculares , Animais , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Neovascularização Patológica , Retina/metabolismo , Doenças Retinianas/metabolismo , Interferon gama/metabolismo , Doenças Vasculares/patologia , Camundongos Endogâmicos C57BL
4.
Immunol Cell Biol ; 100(10): 761-776, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36106449

RESUMO

The role of B-cell-activating factor (BAFF) in B-lymphocyte biology has been comprehensively studied, but its contributions to innate immunity remain unclear. Natural killer (NK) cells form the first line of defense against viruses and tumors, and have been shown to be defective in patients with systemic lupus erythematosus (SLE). The link between BAFF and NK cells in the development and progression of SLE remains unstudied. By assessing NK cell numbers in wild-type (WT), BAFF-/- (BAFF deficient), BAFF-R-/- (BAFF receptor deficient), TACI-/- (transmembrane activator and calcium modulator and cyclophilin ligand interactor deficient), BCMA-/- (B-cell maturation antigen deficient) and BAFF transgenic (Tg) mice, we observed that BAFF signaling through BAFF-R was essential for sustaining NK cell numbers in the spleen. However, according to the cell surface expression of CD27 and CD11b on NK cells, we found that BAFF was dispensable for NK cell maturation. Ex vivo and in vivo models showed that NK cells from BAFF-/- and BAFF Tg mice produced interferon-γ and killed tumor cells at a level similar to that in WT mice. Finally, we established that NK cells do not express receptors that interact with BAFF in the steady state or in the BAFF Tg mouse model of SLE. Our findings demonstrate that BAFF has an indirect effect on NK cell homeostasis and no effect on NK cell function.


Assuntos
Lúpus Eritematoso Sistêmico , Proteína Transmembrana Ativadora e Interagente do CAML , Camundongos , Animais , Proteína Transmembrana Ativadora e Interagente do CAML/genética , Densidade Demográfica , Interleucina-4 , Camundongos Transgênicos , Células Matadoras Naturais/metabolismo
5.
J Mol Biol ; 434(19): 167770, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35907571

RESUMO

The segregation of prokaryotic plasmids typically requires a centromere-like site and two proteins, a centromere-binding protein (CBP) and an NTPase. By contrast, a single 245 residue Par protein mediates partition of the prototypical staphylococcal multiresistance plasmid pSK1 in the absence of an identifiable NTPase component. To gain insight into centromere binding by pSK1 Par and its segregation function we performed structural, biochemical and in vivo studies. Here we show that pSK1 Par binds a centromere consisting of seven repeat elements. We demonstrate this Par-centromere interaction also mediates Par autoregulation. To elucidate the Par centromere binding mechanism, we obtained a structure of the Par N-terminal DNA-binding domain bound to centromere DNA to 2.25 Å. The pSK1 Par structure, which harbors a winged-helix-turn-helix (wHTH), is distinct from other plasmid CBP structures but shows homology to the B. subtilis chromosome segregation protein, RacA. Biochemical studies suggest the region C-terminal to the Par wHTH forms coiled coils and mediates oligomerization. Fluorescence microscopy analyses show that pSK1 Par enhances the separation of plasmids from clusters, driving effective segregation upon cell division. Combined the data provide insight into the molecular properties of a single protein partition system.


Assuntos
Proteínas de Bactérias , Centrômero , Segregação de Cromossomos , Nucleosídeo-Trifosfatase , Plasmídeos , Staphylococcus , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Centrômero/genética , Centrômero/metabolismo , DNA/química , Nucleosídeo-Trifosfatase/metabolismo , Plasmídeos/genética , Staphylococcus/genética
6.
J Clin Immunol ; 42(1): 119-129, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34657245

RESUMO

Rare, biallelic loss-of-function mutations in DOCK8 result in a combined immune deficiency characterized by severe and recurrent cutaneous infections, eczema, allergies, and susceptibility to malignancy, as well as impaired humoral and cellular immunity and hyper-IgE. The advent of next-generation sequencing technologies has enabled the rapid molecular diagnosis of rare monogenic diseases, including inborn errors of immunity. These advances have resulted in the implementation of gene-guided treatments, such as hematopoietic stem cell transplant for DOCK8 deficiency. However, putative disease-causing variants revealed by next-generation sequencing need rigorous validation to demonstrate pathogenicity. Here, we report the eventual diagnosis of DOCK8 deficiency in a consanguineous family due to a novel homozygous intronic deletion variant that caused aberrant exon splicing and subsequent loss of expression of DOCK8 protein. Remarkably, the causative variant was not initially detected by clinical whole-genome sequencing but was subsequently identified and validated by combining advanced genomic analysis, RNA-seq, and flow cytometry. This case highlights the need to adopt multipronged confirmatory approaches to definitively solve complex genetic cases that result from variants outside protein-coding exons and conventional splice sites.


Assuntos
Síndrome de Job , Consanguinidade , Fatores de Troca do Nucleotídeo Guanina/genética , Homozigoto , Humanos , Síndrome de Job/diagnóstico , Síndrome de Job/genética , Mutação/genética , Sequenciamento do Exoma
7.
Sci Rep ; 11(1): 2890, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33536561

RESUMO

We have previously reported evidence that Nogo-A activation of Nogo-receptor 1 (NgR1) can drive axonal dystrophy during the neurological progression of experimental autoimmune encephalomyelitis (EAE). However, the B-cell activating factor (BAFF/BlyS) may also be an important ligand of NgR during neuroinflammation. In the current study we define that NgR1 and its homologs may contribute to immune cell signaling during EAE. Meningeal B-cells expressing NgR1 and NgR3 were identified within the lumbosacral spinal cords of ngr1+/+ EAE-induced mice at clinical score 1. Furthermore, increased secretion of immunoglobulins that bound to central nervous system myelin were shown to be generated from isolated NgR1- and NgR3-expressing B-cells of ngr1+/+ EAE-induced mice. In vitro BAFF stimulation of NgR1- and NgR3-expressing B cells, directed them into the cell cycle DNA synthesis phase. However, when we antagonized BAFF signaling by co-incubation with recombinant BAFF-R, NgR1-Fc, or NgR3 peptides, the B cells remained in the G0/G1 phase. The data suggest that B cells express NgR1 and NgR3 during EAE, being localized to infiltrates of the meninges and that their regulation is governed by BAFF signaling.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/imunologia , Meninges/patologia , Esclerose Múltipla/imunologia , Animais , Linfócitos B/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Meninges/imunologia , Camundongos , Camundongos Knockout , Esclerose Múltipla/patologia , Proteínas Nogo/metabolismo , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Receptores Nogo/metabolismo
8.
Clin Transl Immunology ; 8(12): e01093, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921420

RESUMO

OBJECTIVES: Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease that is difficult to treat. There is currently no optimal stratification of patients with SLE, and thus, responses to available treatments are unpredictable. Here, we developed a new stratification scheme for patients with SLE, based on the computational analysis of patients' whole-blood transcriptomes. METHODS: We applied machine learning approaches to RNA-sequencing (RNA-seq) data sets to stratify patients with SLE into four distinct clusters based on their gene expression profiles. A meta-analysis on three recently published whole-blood RNA-seq data sets was carried out, and an additional similar data set of 30 patients with SLE and 29 healthy donors was incorporated in this study; a total of 161 patients with SLE and 57 healthy donors were analysed. RESULTS: Examination of SLE clusters, as opposed to unstratified SLE patients, revealed underappreciated differences in the pattern of expression of disease-related genes relative to clinical presentation. Moreover, gene signatures correlated with flare activity were successfully identified. CONCLUSION: Given that SLE disease heterogeneity is a key challenge hindering the design of optimal clinical trials and the adequate management of patients, our approach opens a new possible avenue addressing this limitation via a greater understanding of SLE heterogeneity in humans. Stratification of patients based on gene expression signatures may be a valuable strategy allowing the identification of separate molecular mechanisms underpinning disease in SLE. Further, this approach may have a use in understanding the variability in responsiveness to therapeutics, thereby improving the design of clinical trials and advancing personalised therapy.

9.
Immunol Cell Biol ; 96(5): 453-462, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29499091

RESUMO

Altered B-cell homeostasis underlies a wide range of pathologies, from cancers to autoimmunity and immunodeficiency. The molecular safeguards against those disorders, which also allow effective immune responses, are therefore particularly critical. Here, we review recent findings detailing the fine control of B-cell homeostasis, during B-cell development, maturation in the periphery and during activation and differentiation into antibody-producing cells.


Assuntos
Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Doenças do Sistema Imunitário/imunologia , Animais , Diferenciação Celular , Homeostase , Humanos , Tolerância Imunológica , Imunidade Humoral , Ativação Linfocitária
10.
J Autoimmun ; 62: 1-10, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26103922

RESUMO

B cell activating factor of the tumor necrosis factor family (BAFF or BLyS) is a critical factor for B cell survival and maturation. BAFF-transgenic (BAFF-Tg) mice develop autoimmunity that resembles systemic lupus erythematosus (SLE) in a T cell-independent but MyD88-dependent manner, implicating toll-like receptor (TLR) signaling. The specific B cell subtypes that make pro-inflammatory autoantibodies in BAFF-Tg mice are TLR-activated innate B cells known as marginal zone (MZ) and B1 B cells. These cells infiltrate the salivary glands and kidneys of diseased BAFF-Tg mice. However, loss of B1a or MZ B cells does not protect BAFF-Tg mice against disease, suggesting that B1b B cells might be the important pathogenic B cell subset. To test this hypothesis, we have generated BAFF-Tg mice that retained follicular B cells, but are deficient in B1a, B1b and MZ B cells, by crossing BAFF-Tg mice to CD19-deficient mice (BTg-CD19(-/-)). The BTg-CD19(-/-) mice did not produce autoantibodies and were protected from splenomegaly, kidney pathology and all signs of autoimmunity. This work suggests that B1b B cells, rather than MZ or B1a B cells, are sufficient and possibly required for the development of autoimmunity. Loss of the majority of innate-like B cells was able to protect BAFF-Tg mice from developing disease, so we can now conclude that autoimmunity induced by excessive BAFF production requires B1b B cells and CD19 signaling.


Assuntos
Antígenos CD19/genética , Autoimunidade/genética , Autoimunidade/imunologia , Fator Ativador de Células B/metabolismo , Regulação da Expressão Gênica , Animais , Autoanticorpos/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Complemento C3/imunologia , Glomerulonefrite/genética , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Glomerulonefrite/patologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Transdução de Sinais , Receptores Toll-Like/metabolismo
11.
J Autoimmun ; 61: 9-16, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26027434

RESUMO

B cell-activating factor of the TNF family (BAFF) is an essential B cell survival factor. However, high levels of BAFF promote systemic lupus erythematosus (SLE) in mice and humans. Belimumab (anti-human BAFF) limits B cell survival and is approved for use in patients with SLE. Surprisingly, the efficacy of rituximab (anti-human CD20) in SLE remains controversial, despite depleting B cells more potently than belimumab. This raises the question of whether B cell depletion is really the mechanism of action of belimumab. In BAFF transgenic mice, SLE development is T cell-independent but relies on innate activation of B cells via TLRs, and TLR expression is modulated by the BAFF receptor TACI. Here, we show that loss of TACI on B cells protected against BAFF-mediated autoimmune manifestations while preserving B cells, suggesting that loss of BAFF signaling through TACI rather than loss of B cells may underpin the effect of belimumab in the clinic. Therefore, B cell-sparing blockade of TACI may offer a more specific and safer therapeutic alternative to broad B cell depletion in SLE.


Assuntos
Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML/imunologia , Animais , Autoanticorpos/imunologia , Fator Ativador de Células B/genética , Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Citometria de Fluxo , Expressão Gênica/imunologia , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Proteína Transmembrana Ativadora e Interagente do CAML/genética , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo
12.
Exp Eye Res ; 136: 1-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25912997

RESUMO

Oxidative stress is an important contributor to glial and vascular cell damage in ischemic retinopathies. We hypothesized that ebselen via its ability to reduce reactive oxygen species (ROS) and augment nuclear factor-like 2 (Nrf2) anti-oxidants would attenuate hypoxia-induced damage to macroglial Müller cells and also lessen retinal vasculopathy. Primary cultures of rat Müller cells were exposed to normoxia (21% O2), hypoxia (0.5% O2) and ebselen (2.5 µM) for up to 72 h. Oxygen-induced retinopathy (OIR) was induced in C57BL/6J mice while control mice were housed in room air. Mice received vehicle (saline, 5% dimethyl sulfoxide) or ebselen (10 mg/kg) each day between postnatal days 6-18. In cultured Müller cells, flow cytometry for dihydroethidium revealed that ebselen reduced the hypoxia-induced increase in ROS levels, whilst increasing the expression of Nrf2-regulated anti-oxidant genes, heme oxygenase 1, glutathione peroxidase-1, NAD(P)H dehydrogenase quinone oxidoreductase 1 and glutamate-cysteine ligase. Moreover, in Müller cells, ebselen reduced the hypoxia-induced increase in protein levels of pro-angiogenic and pro-inflammatory factors including vascular endothelial growth factor, interleukin-6, monocyte chemoattractant-protein 1 and intercellular adhesion molecule-1, and the mRNA levels of glial fibrillary acidic protein (GFAP), a marker of Müller cell injury. Ebselen improved OIR by attenuating capillary vaso-obliteration and neovascularization and a concomitant reduction in Müller cell gliosis and GFAP. We conclude that ebselen protects against hypoxia-induced injury of retinal Müller cells and the microvasculature, which is linked to its ability to reduce oxidative stress, vascular damaging factors and inflammation. Agents such as ebselen may be potential treatments for retinopathies that feature oxidative stress-mediated damage to glia and the microvasculature.


Assuntos
Antioxidantes/farmacologia , Azóis/farmacologia , Células Ependimogliais/efeitos dos fármacos , Gliose/tratamento farmacológico , Hipóxia/metabolismo , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Degeneração Retiniana/prevenção & controle , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Células Ependimogliais/metabolismo , Citometria de Fluxo , Proteína Glial Fibrilar Ácida , Gliose/metabolismo , Isoindóis , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Degeneração Retiniana/metabolismo , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/prevenção & controle , Vasos Retinianos/efeitos dos fármacos , Superóxidos/metabolismo , Lesões do Sistema Vascular/prevenção & controle
13.
Arterioscler Thromb Vasc Biol ; 34(9): 2033-41, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25012132

RESUMO

OBJECTIVE: Neovascularization and vaso-obliteration are vision-threatening events that develop by interactions between retinal vascular and glial cells. A high-salt diet is causal in cardiovascular and renal disease, which is linked to modulation of the renin-angiotensin-aldosterone system. However, it is not known whether dietary salt influences retinal vasculopathy and if the renin-angiotensin-aldosterone system is involved. We examined whether a low-salt (LS) diet influenced vascular and glial cell injury and the renin-angiotensin-aldosterone system in ischemic retinopathy. APPROACH AND RESULTS: Pregnant Sprague Dawley rats were fed LS (0.03% NaCl) or normal salt (0.3% NaCl) diets, and ischemic retinopathy was induced in the offspring. An LS diet reduced retinal neovascularization and vaso-obliteration, the mRNA and protein levels of the angiogenic factors, vascular endothelial growth factor, and erythropoietin. Microglia, which influence vascular remodeling in ischemic retinopathy, were reduced by LS as was tumor necrosis factor-α. Macroglial Müller cells maintain the integrity of the blood-retinal barrier, and in ischemic retinopathy, LS reduced their gliosis and also vascular leakage. In retina, LS reduced mineralocorticoid receptor, angiotensin type 1 receptor, and renin mRNA levels, whereas, as expected, plasma levels of aldosterone and renin were increased. The aldosterone/mineralocorticoid receptor-sensitive epithelial sodium channel alpha (ENaCα), which is expressed in Müller cells, was increased in ischemic retinopathy and reduced by LS. In cultured Müller cells, high salt increased ENaCα, which was prevented by mineralocorticoid receptor and angiotensin type 1 receptor blockade. Conversely, LS reduced ENaCα, angiotensin type 1 receptor, and mineralocorticoid receptor expression. CONCLUSIONS: An LS diet reduced retinal vasculopathy, by modulating glial cell function and the retinal renin-angiotensin-aldosterone system.


Assuntos
Dieta Hipossódica , Canais Epiteliais de Sódio/fisiologia , Microglia/fisiologia , Sistema Renina-Angiotensina/fisiologia , Neovascularização Retiniana/dietoterapia , Complexo 1 de Proteínas Adaptadoras/análise , Aldosterona/sangue , Aldosterona/fisiologia , Animais , Animais Recém-Nascidos , Aquaporina 4/biossíntese , Aquaporina 4/genética , Peso Corporal , Células Cultivadas , Modelos Animais de Doenças , Comportamento de Ingestão de Líquido , Células Ependimogliais/química , Células Ependimogliais/patologia , Eritropoetina/análise , Gliose/etiologia , Gliose/fisiopatologia , Hematócrito , Transporte de Íons , Isquemia/fisiopatologia , Glomérulos Renais/patologia , Sistema de Sinalização das MAP Quinases , Fosforilação , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Canais de Potássio Corretores do Fluxo de Internalização/genética , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/metabolismo , Neovascularização Retiniana/fisiopatologia , Neovascularização Retiniana/prevenção & controle , Retinopatia da Prematuridade , Sódio/metabolismo , Cloreto de Sódio na Dieta/efeitos adversos , Fator de Necrose Tumoral alfa/biossíntese , Fator A de Crescimento do Endotélio Vascular/análise
14.
Semin Immunol ; 26(3): 191-202, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24996229

RESUMO

Most ligands from the tumour necrosis factor (TNF) superfamily play very important roles in the immune system, and particularly so in B lymphocyte biology. TNF ligands are essential to many aspects of normal B cell biology from development in the bone marrow to maturation in the periphery as well as for activation and differentiation into germinal centre, memory or plasma cells. TNF ligands also influence other aspects of B cell biology such as their ability to present antigens or regulate immune responses. Importantly, inadequate regulation of many TNF ligands is associated with B cell disorders including autoimmunity and cancers. As a result, inhibitors of a number of TNF ligands have been tested in the clinic, with some becoming very successful approved treatments alleviating B cell-mediated pathologies.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Fatores de Necrose Tumoral/metabolismo , Animais , Doenças Autoimunes/imunologia , Linfócitos B/metabolismo , Sobrevivência Celular , Humanos , Linfócitos T/imunologia , Fatores de Necrose Tumoral/classificação
15.
Immunity ; 39(3): 573-83, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24012421

RESUMO

Activation-induced cell death (AICD) plays a critical role in immune homeostasis and tolerance. In T-cell-dependent humoral responses, AICD of B cells is initiated by Fas ligand (FasL) on T cells, stimulating the Fas receptor on B cells. In contrast, T-cell-independent B cell responses involve innate-type B lymphocytes, such as marginal zone (MZ) B cells, and little is known about the mechanisms that control AICD during innate B cell responses to Toll-like receptor (TLR) activation. Here, we show that MZ B cells undergo AICD in response to TLR4 activation in vivo. The transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) receptor and TLR4 cooperate to upregulate expression of both FasL and Fas on MZ B cells and also to repress inhibitors of Fas-induced apoptosis signaling. These findings demonstrate an unappreciated role for TACI and its ligands in the regulation of AICD during T-cell-independent B cell responses.


Assuntos
Apoptose , Proteína Ligante Fas/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Receptor fas/metabolismo , Animais , Receptor do Fator Ativador de Células B/biossíntese , Linfócitos B/imunologia , Ativação Enzimática , Proteína Ligante Fas/biossíntese , Lipopolissacarídeos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Transmembrana Ativadora e Interagente do CAML/genética
16.
Cytokine Growth Factor Rev ; 24(3): 203-15, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23684423

RESUMO

The BAFF system plays a key role in the development of autoimmunity, especially in systemic lupus erythematosus (SLE). This often leads to the assumption that BAFF is mostly a B cell factor with a specific role in autoimmunity. Focus on BAFF and autoimmunity, driven by pharmaceutical successes with the recent approval of a novel targeted therapy Belimumab, has relegated other potential roles of BAFF to the background. Far from being SLE-specific, the BAFF system has a much broader relevance in infection, cancer and allergy. In this review, we provide the latest views on additional roles of the BAFF system in health and diseases, as well as an update on BAFF and autoimmunity, with particular focus on current clinical trials.


Assuntos
Fator Ativador de Células B/fisiologia , Linfócitos B/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Asma/fisiopatologia , Autoimunidade/imunologia , Antígeno de Maturação de Linfócitos B/fisiologia , Infecções Bacterianas/fisiopatologia , Ensaios Clínicos como Assunto , Doença Enxerto-Hospedeiro/fisiopatologia , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Doenças Parasitárias/fisiopatologia , Proteínas Recombinantes de Fusão/uso terapêutico , Proteína Transmembrana Ativadora e Interagente do CAML/fisiologia , Imunologia de Transplantes/fisiologia , Viroses/fisiopatologia
17.
Immunol Rev ; 237(1): 205-25, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20727038

RESUMO

One remarkable feature of the immune system is its capacity to maintain constant numbers of resting immune cells despite the complex nature of signals needed throughout development and maturation. For many years, B-cell survival was thought to rely solely on B-cell receptor (BCR) tonic signals that would trigger necessary basal survival pathways. The discovery of the tumor necrosis factor (TNF)-like ligand BAFF(B-cell activating factor belonging to the TNF family)/BLyS (B-lymphocyte stimulator) changed these views entirely, as BAFF-deficient mice lack most mature B cells, and treatment with BAFF inhibitors leads to their loss, establishing BAFF as an unappreciated key B-cell survival factor. BAFF-mediated survival signals have been mapped and signaling crosstalk with the BCR has been identified, explaining the need for both BCR- and BAFF-mediated signals for B-cell survival. However, this crosstalk only explains how BCR and BAFF signals cooperate to produce survival proteins and yet, inactivating pro-apoptotic factors such as FOXO proteins, which may be managed separately by BAFF and the BCR, has emerged as an equally important step for survival. In this review, we present new views on B-cell survival, at all stages of B-cell life, and suggest that, in most cases, survival results from the production of appropriate survival factors balanced with the adequate and timely degradation of pro-apoptotic proteins.


Assuntos
Fator Ativador de Células B/imunologia , Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais/imunologia , Animais , Autoimunidade/imunologia , Subpopulações de Linfócitos B/citologia , Linfócitos B/citologia , Humanos , Neoplasias/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...