Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Rheumatology (Oxford) ; 63(2): 551-562, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37341646

RESUMEN

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.


Asunto(s)
Nefritis Lúpica , Neutrófilos , Animales , Humanos , Ratones , Leucocitos Mononucleares , Nefritis Lúpica/patología , Neutrófilos/metabolismo , ARN Mensajero/metabolismo , Receptor Toll-Like 7/genética
2.
Nat Commun ; 14(1): 1680, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36973261

RESUMEN

Profiling tumors at single-cell resolution provides an opportunity to understand complexities underpinning lymph-node metastases in head and neck squamous-cell carcinoma. Single-cell RNAseq (scRNAseq) analysis of cancer-cell trajectories identifies a subpopulation of pre-metastatic cells, driven by actionable pathways including AXL and AURK. Blocking these two proteins blunts tumor invasion in patient-derived cultures. Furthermore, scRNAseq analyses of tumor-infiltrating CD8 + T-lymphocytes show two distinct trajectories to T-cell dysfunction, corroborated by their clonal architecture based on single-cell T-cell receptor sequencing. By determining key modulators of these trajectories, followed by validation using external datasets and functional experiments, we uncover a role for SOX4 in mediating T-cell exhaustion. Finally, interactome analyses between pre-metastatic tumor cells and CD8 + T-lymphocytes uncover a putative role for the Midkine pathway in immune-modulation and this is confirmed by scRNAseq of tumors from humanized mice. Aside from specific findings, this study demonstrates the importance of tumor heterogeneity analyses in identifying key vulnerabilities during early metastasis.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Ratones , Animales , Carcinoma de Células Escamosas/patología , Evasión Inmune , Neoplasias de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Linfocitos T CD8-positivos , Linfocitos Infiltrantes de Tumor
3.
Cell Rep ; 41(12): 111841, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36543125

RESUMEN

Neutrophils are critical mediators during the early stages of innate inflammation in response to bacterial or fungal infections. A human hematopoietic system reconstituted in humanized mice aids in the study of human hematology and immunology. However, the poor development of human neutrophils is a well-known limitation of humanized mice. Here, we generate a human granulocyte colony-stimulating factor (hG-CSF) knockin (KI) NOD/Shi-scid-IL2rgnull (NOG) mouse in which hG-CSF is systemically expressed while the mouse G-CSF receptor is disrupted. These mice generate high numbers of mature human neutrophils, which can be readily mobilized into the periphery, compared with conventional NOG mice. Moreover, these neutrophils exhibit infection-mediated emergency granulopoiesis and are capable of efficient phagocytosis and reactive oxygen species production. Thus, hG-CSF KI mice provide a useful model for studying the development of human neutrophils, emergency granulopoiesis, and a potential therapeutic model for sepsis.


Asunto(s)
Mercurio , Neutrófilos , Humanos , Ratones , Animales , Factor Estimulante de Colonias de Granulocitos , Ratones Endogámicos NOD , Hematopoyesis
4.
Curr Protoc ; 1(11): e272, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34748281

RESUMEN

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).


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Discoide , Lupus Eritematoso Sistémico , Animales , Autoanticuerpos , Modelos Animales de Enfermedad , Ratones
5.
Arthritis Rheumatol ; 72(10): 1587-1595, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32524751

RESUMEN

Patients with systemic lupus erythematosus (SLE) display increased numbers of immature neutrophils in the blood, but the exact role of these immature neutrophils is unclear. Neutrophils that sediment within the peripheral blood mononuclear cell fraction after density centrifugation of blood are generally defined as low-density neutrophils (LDNs). Far beyond antimicrobial functions, LDNs are emerging as decision-shapers during innate and adaptive immune responses. Traditionally, neutrophils have been viewed as a homogeneous population. However, the various LDN populations identified in SLE to date are heterogeneously composed of mixed populations of activated mature neutrophils and immature neutrophils at various stages of differentiation. Controversy also surrounds the role of LDNs in SLE in terms of whether they are proinflammatory or polymorphonuclear myeloid-derived suppressor cells. It is clear that LDNs in SLE can secrete increased levels of type I interferon (IFN) and that they contribute to the cycle of inflammation and tissue damage. They readily form neutrophil extracellular traps, exposing modified autoantigens and oxidized mitochondrial DNA, which contribute to autoantibody production and type I IFN signaling, respectively. Importantly, the ability of LDNs in SLE to perform canonical neutrophil functions is polarized, based on mature CD10+ and immature CD10- neutrophils. Although this field is still relatively new, multiomic approaches have advanced our understanding of the diverse origins, phenotype, and function of LDNs in SLE. This review updates the literature on the origin and nature of LDNs, their distinctive features, and their biologic roles in the immunopathogenesis and end-organ damage in SLE.


Asunto(s)
Trampas Extracelulares/metabolismo , Lupus Eritematoso Sistémico/sangre , Neutrófilos/metabolismo , Humanos , Recuento de Leucocitos
6.
Arthritis Res Ther ; 21(1): 185, 2019 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391123

RESUMEN

Following publication of the original article [1], it was brought to our attention that the fifth author's name was incorrectly published. The original article [1] is corrected.

7.
Arthritis Res Ther ; 21(1): 176, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31319876

RESUMEN

OBJECTIVE: This study evaluates the utility of urinary pro-thrombotic molecules such as tissue factor (TF), anti-thrombotic molecules such as tissue factor pathway inhibitor (TFPI), and fibrinolytic molecules such as plasmin and d-dimer as biomarkers of lupus nephritis (LN). METHODS: Urine samples from 113 biopsy-proven LN patients (89 active LN and 24 inactive LN), 45 chronic kidney disease patients, and 41 healthy controls were examined for d-dimer, plasmin, TF, and TFPI levels by ELISA. The area under the receiver operating characteristic curve (AUC) analysis, multivariate regression analysis, and Bayesian network analysis were performed to assess the diagnostic value of the assayed molecules in LN. RESULTS: Although urinary d-dimer, plasmin, TF, and TFPI were all elevated in active LN compared to all control groups, and correlated with rSLEDAI and SLICC RAS disease activity indices, urine plasmin emerged as the strongest independent predictor of eGFR and renal disease status, by multivariate regression analysis and Bayesian network analysis. Whereas urine plasmin discriminated active LN from inactive disease with an AUC of 0.84, the combination of urine plasmin and TFPI discriminated ALN from ILN with an AUC of 0.86, with both surpassing the specificity and positive predictive value of traditional markers such as anti-dsDNA and complement C3. CONCLUSION: Both thrombogenic and thrombolytic cascades appear to be upregulated in lupus nephritis, with proteins from both cascades appearing in the urine. Of the coagulation cascade proteins surveyed, urine plasmin emerges as the strongest predictor of eGFR and clinical renal disease in patients with LN.


Asunto(s)
Biomarcadores/orina , Productos de Degradación de Fibrina-Fibrinógeno/orina , Fibrinolisina/orina , Lipoproteínas/orina , Nefritis Lúpica/orina , Tromboplastina/orina , Adulto , Teorema de Bayes , Femenino , Humanos , Nefritis Lúpica/diagnóstico , Masculino , Persona de Mediana Edad , Análisis Multivariante , Análisis de Regresión , Sensibilidad y Especificidad , Adulto Joven
8.
Front Immunol ; 10: 1546, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31354711

RESUMEN

The global increase in autoimmunity, together with the emerging autoimmune-related side effects of cancer immunotherapy, have furthered a need for understanding of immune tolerance and activation. Systemic lupus erythematosus (SLE) is the archetypical autoimmune disease, affecting multiple organs, and tissues. Studying SLE creates knowledge relevant not just for autoimmunity, but the immune system in general. Murine models and patient studies have provided increasing evidence for the innate immune toll like receptor-7 (TLR7) in disease initiation and progression. Here, we demonstrated that the kinase activity of the TLR7-downstream signaling molecule, interleukin-1 receptor associated kinase 4 (IRAK4), is essential for mild and severe autoimmune traits of the Sle1 and Sle1-TLR7 transgenic (Sle1Tg7) murine models, respectively. Elimination of IRAK4 signaling prevented all pathological traits associated with murine lupus, including splenomegaly with leukocyte expansion, detectable circulating antinuclear antibodies and glomerulonephritis, in both Sle1 and Sle1Tg7 mice. The expansion of germinal center B cells and increased effector memory T cell phenotypes that are typical of lupus-prone strains, were also prevented with IRAK4 kinase elimination. Analysis of renal leukocyte infiltrates confirmed our earlier findings of an expanded conventional dendritic cell (cDC) within the kidneys of nephritic mice, and this was prevented with IRAK4 kinase elimination. Analysis of TLR7 at the protein level revealed that the expression in immune cells is dependent on the TLR7-transgene itself and/or autoimmune disease factors in a cell-specific manner. Increased TLR7 protein expression in renal macrophages and cDCs correlated with disease parameters such as blood urea nitrogen (BUN) levels and the frequency of leukocytes infiltrating the kidney. These findings suggest that controlling the level of TLR7 or downstream signaling within myeloid populations may prevent chronic inflammation and severe nephritis.


Asunto(s)
Células Dendríticas/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Riñón/patología , Leucocitos/fisiología , Lupus Eritematoso Sistémico/metabolismo , Nefritis Lúpica/metabolismo , Macrófagos/inmunología , Receptor Toll-Like 7/metabolismo , Animales , Anticuerpos Antinucleares/sangre , Movimiento Celular , Modelos Animales de Enfermedad , Glomerulonefritis , Humanos , Inmunidad Innata , Quinasas Asociadas a Receptores de Interleucina-1/genética , Riñón/metabolismo , Nefritis Lúpica/genética , Ratones , Ratones Transgénicos , Especificidad de Órganos , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Transducción de Señal , Receptor Toll-Like 7/genética
9.
Arthritis Rheumatol ; 70(10): 1597-1609, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29687651

RESUMEN

OBJECTIVE: Toll-like receptors (TLRs) 7 and 9 are important innate signaling molecules with opposing roles in the development and progression of systemic lupus erythematosus (SLE). While multiple studies support the notion of a dependency on TLR-7 for disease development, genetic ablation of TLR-9 results in severe disease with glomerulonephritis (GN) by a largely unknown mechanism. This study was undertaken to examine the suppressive role of TLR-9 in the development of severe lupus in a mouse model. METHODS: We crossed Sle1 lupus-prone mice with TLR-9-deficient mice to generate Sle1TLR-9-/- mice. Mice ages 4.5-6.5 months were evaluated for severe autoimmunity by assessing splenomegaly, GN, immune cell populations, autoantibody and total Ig profiles, kidney dendritic cell (DC) function, and TLR-7 protein expression. Mice ages 8-10 weeks were used for functional B cell studies, Ig profiling, and determination of TLR-7 expression. RESULTS: Sle1TLR-9-/- mice developed severe disease similar to TLR-9-deficient MRL and Nba2 models. Sle1TLR-9-/- mouse B cells produced more class-switched antibodies, and the autoantibody repertoire was skewed toward RNA-containing antigens. GN in these mice was associated with DC infiltration, and purified Sle1TLR-9-/- mouse renal DCs were more efficient at TLR-7-dependent antigen presentation and expressed higher levels of TLR-7 protein. Importantly, this increase in TLR-7 expression occurred prior to disease development, indicating a role in the initiation stages of tissue destruction. CONCLUSION: The increase in TLR-7-reactive immune complexes, and the concomitant enhanced expression of their receptor, promotes inflammation and disease in Sle1TLR9-/- mice.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/deficiencia , Regulación hacia Arriba/inmunología , Animales , Antígenos/inmunología , Modelos Animales de Enfermedad , Ratones , ARN/inmunología , Receptor Toll-Like 9/inmunología
10.
Rheumatology (Oxford) ; 56(suppl_1): i55-i66, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28375453

RESUMEN

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.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Citocinas/inmunología , Ambiente , Inflamación/inmunología , Lupus Eritematoso Sistémico/inmunología , Autoinmunidad/genética , Predisposición Genética a la Enfermedad , Humanos , Inflamación/genética , Lupus Eritematoso Sistémico/genética , Autotolerancia/genética , Autotolerancia/inmunología
11.
Rheumatology (Oxford) ; 56(suppl_1): i88-i99, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28013204

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/inmunología , Ratones , Animales , Humanos , Lupus Eritematoso Sistémico/genética
12.
Int Immunol ; 28(5): 223-32, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26567289

RESUMEN

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.


Asunto(s)
Presentación de Antígeno , Macrófagos/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/inmunología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Ligandos , Lipopolisacáridos/toxicidad , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética
13.
Proc Natl Acad Sci U S A ; 112(45): E6195-204, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26512111

RESUMEN

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.


Asunto(s)
Células Dendríticas/metabolismo , Nefritis Lúpica/fisiopatología , Receptor Toll-Like 7/metabolismo , Regulación hacia Arriba , Análisis de Varianza , Animales , Secuencia de Bases , Antígeno CD11b/metabolismo , Cartilla de ADN/genética , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Humanos , Procesamiento de Imagen Asistido por Computador , Glomérulos Renales/citología , Glomérulos Renales/patología , Nefritis Lúpica/metabolismo , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Estadísticas no Paramétricas
14.
Front Pharmacol ; 5: 265, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25538618

RESUMEN

Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by the loss of tolerance to self-nuclear antigens. The symptoms of SLE, progression of pathology and the array of autoantibodies present in the serum differ significantly from patient to patient, which calls for a personalized approach to treatment. SLE is polygenic and strongly influenced by gender, ethnicity, and environmental factors. Data from genome-wide association studies suggests that polymorphisms in as many as 100 genes contribute to SLE susceptibility. Recent research has focused on genes associated with Toll-like receptors (TLRs), type I interferons, immune regulation pathways, and immune-complex clearance. TLR7 and TLR9 have been extensively studied using lupus-prone mouse models. In multiple systems overexpression of TLR7 drives disease progression but interestingly, a loss of TLR9 results in an almost identical phenotype. While TLR7 overexpression has been linked to human SLE, the possible role of TLR9 in human disease remains elusive. In the present review, we focus on TLR polymorphisms and TLR expression in SLE patients and discuss their potential as biomarkers for individualized treatment.

15.
J Immunol ; 191(12): 6128-35, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24218451

RESUMEN

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.


Asunto(s)
Exodesoxirribonucleasas/fisiología , Inflamación/inmunología , Activación de Macrófagos/fisiología , Fosfoproteínas/fisiología , Animales , Presentación de Antígeno , Apoptosis , Antígeno B7-2/biosíntesis , Antígeno B7-2/genética , Química Encefálica , Exodesoxirribonucleasas/deficiencia , Exodesoxirribonucleasas/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/metabolismo , Interferón-alfa/biosíntesis , Interferón-alfa/genética , Células Jurkat , Células L , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fagocitosis , Fosfoproteínas/deficiencia , Fosfoproteínas/inmunología , Proteínas Recombinantes/farmacología , Linfocitos T/inmunología , Linfocitos T/patología , Receptor Toll-Like 9/fisiología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
16.
J Pharm Pharmacol ; 64(8): 1090-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22775212

RESUMEN

OBJECTIVES: Delineation of EP4 receptor signalling properties in immature B cells. METHODS: WEHI 231 cells were used as a model of immature B lymphocytes. The effects of PGE2, EP4 receptor antagonist, EP4 receptor agonist, forskolin and adenylate cyclase inhibitor on proliferation of WEHI 231 cells were examined by MTS assay. Cyclic adenosine monophosphate (cAMP) levels were examined by ELISA, whereas phosphorylation of vasodilator-stimulated phosphoprotein (VASP), kinase, extracellular signal-regulated kinase1/2, IκB-α and nuclear factor (NF)-κB subunit p105 were subjected to Western blot analysis. Translocation of NF-κB subunit p65 and EPRAP (EP4 receptor associated protein) was examined by fluorescence microscopy. Levels of early growth response factor (Egr)-1 mRNA were determined by quantitative PCR. KEY FINDINGS: We identified the EP4 receptor as the principal molecule mediating the growth-suppressive effect of prostaglandin E2 in WEHI 231 cells. EP4 receptor activation results in cAMP formation and the activation of protein kinase A, NF-κB1 p105 subunit stabilization and inhibition of IκBα phosphorylation, followed by the accumulation of NF-κB p65 subunit in the cell cytoplasm, whereas the activation of PI3K is not involved in EP4 receptor signalling. Elevation of cAMP and inhibition of NF-κB activation are two possible mechanisms by which the EP4 receptor inhibits the proliferation of immature B lymphocytes. CONCLUSIONS: Modulation of the EP4 receptor on immature B lymphocytes provides important insight into the observed action of PGE2 and opens new possibilities for the development of therapies for autoimmune diseases, leukaemia and lymphomas.


Asunto(s)
Proliferación Celular , AMP Cíclico/metabolismo , Dinoprostona/metabolismo , FN-kappa B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Inhibidores de Adenilato Ciclasa , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas I-kappa B/metabolismo , Enfermedades del Sistema Inmune/tratamiento farmacológico , Ratones , Inhibidor NF-kappaB alfa , Transducción de Señal
17.
Prostaglandins Other Lipid Mediat ; 95(1-4): 19-26, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21600299

RESUMEN

Prostaglandin E2 (PGE2) is emerging as an important co-modulator of B cell responses. Using a pharmacological approach, we aimed to delineate the role of PGE2 in B cell receptor (BCR) induced apoptosis of immature B cells. Gene and protein expression analyses showed that, of the four PGE2 receptors subtypes, only EP4 receptor is upregulated upon BCR cross-linking, leading to sensitization of WEHI 231 cells towards PGE2 mediated inhibitory effects. EP4 receptor antagonist ONO-AE3-208, was able to completely revert the observed effects of PGE2. The engagement of EP4 receptor promotes BCR-induced G0/G1 arrest of WEHI 231 cells, resulting in enhanced caspase mediated, BCR-induced apoptosis. We addressed, mechanistically, the interplay between BCR and EP4 receptor signaling components. Prostaglandin1-alcohol (Pge1-OH), a selective EP4 receptor agonist inhibits BCR-induced activation of NF-κB by suppression of BCR-induced IκBα phosphorylation. Disruption of prosurvival pathways is a possible mechanism by which PGE2 enhances BCR-induced apoptosis in immature B lymphocytes.


Asunto(s)
Apoptosis , Receptores de Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/fisiología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dinoprostona/farmacología , Dinoprostona/fisiología , Expresión Génica , Interfase , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Naftalenos/farmacología , Fenilbutiratos/farmacología , Células Precursoras de Linfocitos B/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/genética
18.
Eur J Pharmacol ; 602(1): 15-22, 2009 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19032950

RESUMEN

Molecules regulating cell death constitute prominent therapeutic targets. The pro-apoptotic role of serine protease inhibitors prompted us to search for novel modulators of this process. We have tested some recently synthesized antithrombotic compounds for their potential to induce apoptotic cell death. Cell based analyses revealed that inhibitors built on the azaphenylalanine scaffold are, for B-cell lymphoma cells, severely cytotoxic, while other compounds tested were moderate or non-cytotoxic. These inhibitors induced the time and concentration dependent biochemical and morphological characteristics of apoptosis, such as DEVDase activation, loss of mitochondrial membrane potential, nuclear degradation and genomic DNA fragmentation. Most of the inhibitors proved to be selective for thrombin, with inhibition constants (K(i)) in the nanomolar range. However, they could also inhibit at least one additional serine protease (trypsin, chymotrypsin and/or coagulation factor X) with K(i) values in the nanomolar or low micromolar range. These serine protease inhibitors constitute novel apoptosis inducing compounds in B-cell lymphoma cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Fenilalanina/análogos & derivados , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Línea Celular Tumoral , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Fenilalanina/farmacología , Factores de Tiempo
19.
Cancer Genet Cytogenet ; 187(1): 28-33, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18992638

RESUMEN

The involvement of the CYP11A1 gene in the synthesis of androgens makes it a compelling candidate for various hormone-dependent diseases, including prostate cancer. A microsatellite polymorphism (TTTTA)n in the promoter region of the CYP11A1 gene has been reported to be associated with an increased risk of metastatic and high-grade prostate cancer. In the present study of 110 prostate cancer patients and 96 population controls, we examined the association between the CYP11A1 (TTTTA)n polymorphism and prostate cancer risk, aggressiveness, and incidence of biochemical relapse after prostatectomy. We have also evaluated the potential of the (TTTTA)n polymorphism as a microsatellite marker for the detection of genomic instability in prostate cancer. A strong association of the genotype containing the (TTTTA)6 allele with the occurrence of biochemical relapse after prostatectomy in patients with organ confined prostate cancer (p<0.0001), as well as in patients with low-grade prostate cancer (p=0.002) or both (p<0.0003) was determined. The incidence of biochemical relapse in patients with organ confined and low-grade prostate cancer in our study group was 22%, but increased to 50% in carriers of the (TTTTA)6 allele. Our findings also suggest (TTTTA)n instability as a potential marker for the detection of early events in carcinogenesis.


Asunto(s)
Alelos , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Neoplasias de los Genitales Masculinos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Vesículas Seminales/patología , Adolescente , Adulto , Secuencia de Bases , Genotipo , Humanos , Masculino , Repeticiones de Microsatélite/genética , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Recurrencia , Valores de Referencia , Factores de Riesgo , Eslovenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...