Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 201(3): 940-949, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950509

RESUMEN

Central tolerance checkpoints are critical for the elimination of autoreactive B cells and the prevention of autoimmunity. When autoreactive B cells encounter their Ag at the immature B cell stage, BCR cross-linking induces receptor editing, followed by apoptosis if edited cells remain autoreactive. Although the transcription factor Foxo1 is known to promote receptor editing, the role of the related factor Foxo3 in central B cell tolerance is poorly understood. We find that BCR-stimulated immature B cells from Foxo3-deficient mice demonstrate reduced apoptosis compared with wild type cells. Despite this, Foxo3-/- mice do not develop increased autoantibodies. This suggests that the increased survival of Foxo3-/- immature B cells allows additional rounds of receptor editing, resulting in more cells "redeeming" themselves by becoming nonautoreactive. Indeed, increased Igλ usage and increased recombining sequence recombination among Igλ-expressing cells were observed in Foxo3-/- mice, indicative of increased receptor editing. We also observed that deletion of high-affinity autoreactive cells was intact in the absence of Foxo3 in the anti-hen egg lysozyme (HEL)/membrane-bound HEL model. However, Foxo3 levels in B cells from systemic lupus erythematosus (SLE) patients were inversely correlated with disease activity and reduced in patients with elevated anti-dsDNA Abs. Although this is likely due in part to increased B cell activation in these SLE patients, it is also possible that low-affinity B cells that remain autoreactive after editing may survive inappropriately in the absence of Foxo3 and become activated to secrete autoantibodies in the context of other SLE-associated defects.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , Proteína Forkhead Box O3/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Autoinmunidad/inmunología , Diferenciación Celular/inmunología , Femenino , Tolerancia Inmunológica/inmunología , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Precursoras de Linfocitos B/inmunología
2.
J Immunol ; 196(4): 1541-9, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26764035

RESUMEN

The contribution of autoantibody-producing plasma cells in multiple sclerosis (MS) remains unclear. Anti-CD20 B cell depletion effectively reduces disease activity in MS patients, but it has a minimal effect on circulating autoantibodies and oligoclonal bands in the cerebrospinal fluid. Recently we reported that MEDI551, an anti-CD19 mAb, therapeutically ameliorates experimental autoimmune encephalomyelitis (EAE), the mouse model of MS. MEDI551 potently inhibits pathogenic adaptive immune responses, including depleting autoantibody-producing plasma cells. In the present study, we demonstrated that CD19 mAb treatment ameliorates EAE more effectively than does CD20 mAb. Myelin oligodendrocyte glycoprotein-specific Abs and short-lived and long-lived autoantibody-secreting cells were nearly undetectable in the CD19 mAb-treated mice, but they remained detectable in the CD20 mAb-treated mice. Interestingly, residual disease severity in the CD20 mAb-treated animals positively correlated with the frequency of treatment-resistant plasma cells in the bone marrow. Of note, treatment-resistant plasma cells contained a substantial proportion of CD19(+)CD20(-) plasma cells, which would have otherwise been targeted by CD19 mAb. These data suggested that CD19(+)CD20(-) plasma cells spared by anti-CD20 therapy likely contribute to residual EAE severity by producing autoreactive Abs. In patients with MS, we also identified a population of CD19(+)CD20(-) B cells in the cerebrospinal fluid that would be resistant to CD20 mAb treatment.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD19/inmunología , Antígenos CD20/inmunología , Linfocitos B/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Células Plasmáticas/inmunología , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Autoanticuerpos/análisis , Líquido Cefalorraquídeo/química , Líquido Cefalorraquídeo/citología , Líquido Cefalorraquídeo/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología
3.
Rheumatology (Oxford) ; 56(suppl_1): i100-i113, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28375452

RESUMEN

This review summarizes traditional and emerging therapies for SLE. Evidence suggests that the heterogeneity of SLE is a crucial aspect contributing to the failure of large clinical trials for new targeted therapies. A clearer understanding of the mechanisms driving disease pathogenesis combined with recent advances in medical science are predicted to enable accelerated progress towards improved SLE diagnosis and personalized approaches to treatment.


Asunto(s)
Antirreumáticos/uso terapéutico , Glucocorticoides/uso terapéutico , Inmunosupresores/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Azatioprina/uso terapéutico , Ciclofosfamida/uso terapéutico , Descubrimiento de Drogas , Humanos , Hidroxicloroquina/uso terapéutico , Factores Inmunológicos/uso terapéutico , Interleucina-2/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Metotrexato/uso terapéutico , Terapia Molecular Dirigida , Ácido Micofenólico/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Rituximab , Ustekinumab/uso terapéutico , Vitamina D/uso terapéutico
4.
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
5.
Clin Immunol ; 169: 58-68, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27237127

RESUMEN

Systemic lupus erythematosus (SLE) is characterized by antibody-mediated chronic inflammation in the kidney, lung, skin, and other organs to cause inflammation and damage. Several inflammatory pathways are dysregulated in SLE, and understanding these pathways may improve diagnosis and treatment. In one such pathway, Axl tyrosine kinase receptor responds to Gas6 ligand to block inflammation in leukocytes. A soluble form of the Axl receptor ectodomain (sAxl) is elevated in serum from patients with SLE and lupus-prone mice. We hypothesized that sAxl in SLE serum originates from the surface of leukocytes and that the loss of leukocyte Axl contributes to the disease. We determined that macrophages and B cells are a source of sAxl in SLE and in lupus-prone mice. Shedding of the Axl ectodomain from the leukocytes of lupus-prone mice is mediated by the matrix metalloproteases ADAM10 and TACE (ADAM17). Loss of Axl from lupus-prone macrophages renders them unresponsive to Gas6-induced anti-inflammatory signaling in vitro. This phenotype is rescued by combined ADAM10/TACE inhibition. Mice with Axl-deficient macrophages develop worse disease than controls when challenged with anti-glomerular basement membrane (anti-GBM) sera in an induced model of nephritis. ADAM10 and TACE also mediate human SLE PBMC Axl cleavage. Collectively, these studies indicate that increased metalloprotease-mediated cleavage of leukocyte Axl may contribute to end organ disease in lupus. They further suggest dual ADAM10/TACE inhibition as a potential therapeutic modality in SLE.


Asunto(s)
Proteína ADAM10/inmunología , Proteína ADAM17/inmunología , Lupus Eritematoso Sistémico/inmunología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Adulto , Animales , Western Blotting , Línea Celular , Femenino , Expresión Génica/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Noqueados , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven , Tirosina Quinasa del Receptor Axl
6.
Cytokine ; 73(2): 236-44, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25794663

RESUMEN

The cytokines IL-6 and IL-10 are produced by cells of the adaptive and innate arms of the immune system and they appear to play key roles in genetically diverse autoimmune diseases such as relapsing remitting multiple sclerosis (MS), rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Whereas previous intense investigations focused on the generation of autoantibodies and their contribution to immune-mediated pathogenesis in these diseases; more recent attention has focused on the roles of cytokines such as IL-6 and IL-10. In response to pathogens, antigen presenting cells (APC), including B cells, produce IL-6 and IL-10 in order to up-or down-regulate immune cell activation and effector responses. Evidence of elevated levels of the proinflammatory cytokine IL-6 has been routinely observed during inflammatory responses and in a number of autoimmune diseases. Our recent studies suggest that MS peripheral blood B cells secrete higher quantities of IL-6 and less IL-10 than B cells from healthy controls. Persistent production of IL-6, in turn, contributes to T cell expansion and the functional hyperactivity of APC such as MS B cells. Altered B cell activity can have a profound impact on resultant T cell effector functions. Enhanced signaling through the IL-6 receptor can effectively inhibit cytolytic activity, induce T cell resistance to IL-10-mediated immunosuppression and increase skewing of autoreactive T cells to a pathogenic Th17 phenotype. Our recent findings and studies by others support a role for the indirect attenuation of B cell responses by Glatiramer acetate (GA) therapy. Our studies suggest that GA therapy temporarily permits homeostatic regulatory mechanisms to be reinstated. Future studies of mechanisms underlying dysregulated B cell cytokine production could lead to the identification of novel targets for improved immunoregulatory therapies for autoimmune diseases.


Asunto(s)
Autoinmunidad , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Esclerosis Múltiple/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Inmunomodulación , Esclerosis Múltiple/genética , Esclerosis Múltiple/terapia
7.
Purinergic Signal ; 11(3): 347-59, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26054298

RESUMEN

Recruitment of monocytes in the liver is a key pathogenic feature of hepatic inflammation in nonalcoholic steatohepatitis (NASH), but the mechanisms involved are poorly understood. Here, we studied migration of human monocytes in response to supernatants obtained from liver cells after inducing lipoapoptosis with saturated free fatty acids (FFA). Lipoapoptotic supernatants stimulated monocyte migration with the magnitude similar to a monocyte chemoattractant protein, CCL2 (MCP-1). Inhibition of c-Jun NH2-terminal kinase (JNK) in liver cells with SP600125 blocked migration of monocytes in a dose-dependent manner, indicating that JNK stimulates release of chemoattractants in lipoapoptosis. Notably, treatment of supernatants with Apyrase to remove ATP potently inhibited migration of THP-1 monocytes and partially blocked migration of primary human monocytes. Inhibition of the CCL2 receptor (CCR2) on THP-1 monocytes with RS102895, a specific CCR2 inhibitor, did not block migration induced by lipoapoptotic supernatants. Consistent with these findings, lipoapoptosis stimulated pathophysiological extracellular ATP (eATP) release that increased supernatant eATP concentration from 5 to ~60 nM. Importantly, inhibition of Panx1 expression in liver cells with short hairpin RNA (shRNA) decreased supernatant eATP concentration and inhibited monocyte migration, indicating that monocyte migration is mediated in part by Panx1-dependent eATP release. Moreover, JNK inhibition decreased supernatant eATP concentration and inhibited Pannexin1 activation, as determined by YoPro-1 uptake in liver cells in a dose-dependent manner. These results suggest that JNK regulates activation of Panx1 channels, and provide evidence that Pannexin1-dependent pathophysiological eATP release in lipoapoptosis is capable of stimulating migration of human monocytes, and may participate in the recruitment of monocytes in chronic liver injury induced by saturated FFA.


Asunto(s)
Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Conexinas/fisiología , Ácidos Grasos no Esterificados/farmacología , Ácidos Grasos/farmacología , Hígado/metabolismo , Monocitos/efectos de los fármacos , Proteínas del Tejido Nervioso/fisiología , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Quimiocina CCL2/antagonistas & inhibidores , Conexinas/genética , Humanos , Hígado/citología , Hígado/efectos de los fármacos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Proteínas del Tejido Nervioso/genética , Ácido Palmítico/farmacología , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Ratas , Receptores CCR2/antagonistas & inhibidores
8.
Blood ; 118(14): 3890-900, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21832277

RESUMEN

CD8(+) cytotoxic T lymphocytes play a major role in defense against intracellular pathogens, and their functions are specified by antigen recognition and innate cytokines. IL-12 and IFN-α/ß are potent "signal 3" cytokines that are involved in both effector and memory cell development. Although the majority of effector cells are eliminated as inflammation resolves, some survive within the pool of memory cells and retain immediate effector function. In this study, we demonstrate that IL-12 instructs a unique program of effector cell differentiation that is distinct from IFN-α/ß. Moreover, effector memory (T(EM)) cells within peripheral blood display many common attributes of cells differentiated in vitro in response to IL-12, including proinflammatory cytokine secretion and lytic activity. A pattern of IL-12-induced genes was identified that demarcate T(EM) from central memory cells, and the ontologies of these genes correlated precisely with their effector functions. Further, we uncovered a unique program of gene expression that was acutely regulated by IL-12 and reflected in stable gene expression patterns within T(EM), but not T central memory cells in vivo. Thus, this study directly links a selective set of IL-12-induced genes to the programming of effector functions within the stable population of human CD8(+) T(EM) cells in vivo.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Regulación de la Expresión Génica , Interferón Tipo I/inmunología , Interleucina-12/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Citocinas/inmunología , Humanos , Memoria Inmunológica , Receptores CCR7/genética , Receptores CCR7/inmunología , Receptores CXCR3/genética , Receptores CXCR3/inmunología
9.
Eur J Immunol ; 40(10): 2942-56, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20812237

RESUMEN

Recent evidence suggests that B- and T-cell interactions may be paramount in relapsing-remitting MS (RRMS) disease pathogenesis. We hypothesized that memory B-cell pools from RRMS patients may specifically harbor a subset of potent neuro-APC that support neuro-Ag reactive T-cell proliferation and cytokine secretion. To test this hypothesis, we compared CD80 and HLA-DR expression, IL-10 and lymphotoxin-α secretion, neuro-Ag binding capacity, and neuro-Ag presentation by memory B cells from RRMS patients to naïve B cells from RRMS patients and to memory and naïve B cells from healthy donors (HD). We identified memory B cells from some RRMS patients that elicited CD4(+) T-cell proliferation and IFN-γ secretion in response to myelin basic protein and myelin oligodendrocyte glycoprotein. Notwithstanding the fact that the phenotypic parameters that promote efficient Ag presentation were observed to be similar between RRMS and HD memory B cells, a corresponding capability to elicit CD4(+) T-cell proliferation in response to myelin basic protein and myelin oligodendrocyte glycoprotein was not observed in HD memory B cells. Our results demonstrate for the first time that the memory B-cell pool in RRMS harbors neuro-Ag specific B cells that can activate T cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica/inmunología , Interferón gamma/biosíntesis , Proteína Básica de Mielina/inmunología , Glicoproteína Asociada a Mielina/inmunología , Adulto , Estudios de Cohortes , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Interferón gamma/sangre , Interferón gamma/inmunología , Activación de Linfocitos , Linfotoxina-alfa/inmunología , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/inmunología , Proteínas de la Mielina , Glicoproteína Mielina-Oligodendrócito , Adulto Joven
10.
Arthritis Rheum ; 62(11): 3436-46, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20722038

RESUMEN

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.


Asunto(s)
Quimiocina CXCL12/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Receptores CXCR4/metabolismo , Adolescente , Adulto , Anciano , Quimiocina CXCL12/inmunología , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Leucocitos/inmunología , Leucocitos/metabolismo , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Masculino , Persona de Mediana Edad , Receptores CXCR4/inmunología , Regulación hacia Arriba/inmunología
11.
Clin Mol Allergy ; 9: 12, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21961521

RESUMEN

BACKGROUND: To examine whether whole genome expression profiling could reveal changes in mRNA expression of peripheral blood mononuclear cells (PBMC) from allergic patients undergoing rush immunotherapy (RIT) that might be manifest within the first few months of treatment. METHODS: For this study, PBMC from three allergic patients undergoing RIT were assessed at four timepoints: prior to RIT, at 1 week and 7 week post-RIT, during build-up and at 4 months, after establishment of a maintenance dose. PBMC mRNA gene expression changes over time were determined by oligonucleotide microarrays using the Illumina Human-6 BeadChip Platform, which simultaneously interrogates expression profiles of > 47,000 transcripts. Differentially expressed genes were identified using well-established statistical analysis for microarrays. In addition, we analyzed peripheral blood basophil high-affinity IgE receptor (Fc epsilon RI) expression and T-regulatory cell frequency as detected by expression of CD3+CD4+CD25bright cells at each timepoint using flow cytometry. RESULTS: In comparing the initial 2 timepoints with the final 2 timepoints and analyzing for genes with ≥1.5-fold expression change (p less than or equal to 0.05, BH-FDR), we identified 507 transcripts. At a 2-fold change (p less than or equal to 0.05, BH-FDR), we found 44 transcripts. Of these, 28 were up-regulated and 16 were down-regulated genes. From these datasets, we have identified changes in immunologically relevant genes from both the innate and adaptive response with upregulation of expressed genes for molecules including IL-1ß, IL-8, CD40L, BTK and BCL6. At the 4 month timepoint, we noted a downward trend in Fc epsilon RI expression in each of the three patients and increased allergen-specific IgG4 levels. No change was seen in the frequency of peripheral T-regulatory cells expressed over the four timepoints. CONCLUSIONS: We observed significant changes in gene expression early in peripheral blood samples from allergic patients undergoing RIT. Moreover, serum levels for allergen specific IgG4 also increased over the course of treatment. These studies suggest that RIT induces rapid and dynamic alterations in both innate and adaptive immunity which can be observed in the periphery of allergic patients. These alterations could be directly related to the therapeutic shift in the allergen-specific class of immunoglobulin.

12.
J Clin Invest ; 117(8): 2186-96, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17641780

RESUMEN

Though B cells play key roles in lupus pathogenesis, the molecular circuitry and its dysregulation in these cells as disease evolves remain poorly understood. To address this, a comprehensive scan of multiple signaling axes using multiplexed Western blotting was undertaken in several different murine lupus strains. PI3K/AKT/mTOR (mTOR, mammalian target of rapamycin), MEK1/Erk1/2, p38, NF-kappaB, multiple Bcl-2 family members, and cell-cycle molecules were observed to be hyperexpressed in lupus B cells in an age-dependent and lupus susceptibility gene-dose-dependent manner. Therapeutic targeting of the AKT/mTOR axis using a rapamycin (sirolimus) derivative ameliorated the serological, cellular, and pathological phenotypes associated with lupus. Surprisingly, the targeting of this axis was associated with the crippling of several other signaling axes. These studies reveal that lupus pathogenesis is contingent upon the activation of an elaborate network of signaling cascades that is shared among genetically distinct mouse models and raise hope that targeting pivotal nodes in these networks may offer therapeutic benefit.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Transducción de Señal/inmunología , Factores de Edad , Animales , Linfocitos B/patología , Modelos Animales de Enfermedad , Dosificación de Gen/inmunología , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/inmunología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirolimus/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR
13.
J Immunol ; 181(12): 8204-8, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050236

RESUMEN

IL-2 is a hallmark cytokine secreted by central memory CD4(+) T cells (T(CM)). Although naive cells rapidly secrete IL-2 in response to Ag stimulation, IL-12 inhibits IL-2 secretion in daughter cells as they differentiate into Th1 cells. In this study, we uncover a unique role for IFN-alpha in regulating IL-2 secretion by human T(CM) cells. IFN-alpha synergized with IL-12 to enhance a subset of cells that secreted high and sustained levels of IL-2. These IL-2-secreting cells displayed phenotypic and functional characteristics of T(CM) and were capable of generating IFN-gamma-secreting effectors upon secondary activation. T-bet has been implicated in negatively regulating IL-2 secretion in murine T cells; however, T-bet expression did not inhibit IFN-alpha-dependent IL-2 secretion in human T(CM) cells. Thus, our results highlight a unique role for IFN-alpha in regulating the development of IL-2-secreting human T(CM) cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Memoria Inmunológica , Interferón-alfa/fisiología , Interleucina-2/metabolismo , Proteínas de Dominio T Box/fisiología , Subgrupos de Linfocitos T/metabolismo , Adulto , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Interferón beta/fisiología , Interleucina-12/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Células TH1/citología , Células TH1/inmunología
14.
Sci Immunol ; 3(20)2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29429981

RESUMEN

Gut bacteria influence patient response to cancer therapy.


Asunto(s)
Bacterias , Neoplasias , Sistema Digestivo , Tracto Gastrointestinal/microbiología , Humanos , Inmunoterapia , Intestinos , Filogenia
15.
Sci Immunol ; 2(14)2017 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-28778906

RESUMEN

A transancestral SLE study examining the relationship between genetic load and disease risk.

16.
Sci Immunol ; 2(17)2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29101211

RESUMEN

TNF and type I interferons alter TLR4 responses by reprogramming the macrophage genome.


Asunto(s)
Citocinas , Interferón Tipo I , Macrófagos , Yin-Yang
17.
Sci Immunol ; 2(11)2017 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-28763792

RESUMEN

The long noncoding RNA Flicr modifies Foxp3 chromatin accessibility to reduce T regulatory cell differentiation and function.

18.
Exp Ther Med ; 13(5): 2183-2192, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28565826

RESUMEN

Anti-nuclear antibodies (ANAs) may be induced in patients with rheumatoid arthritis (RA) receiving anti-tumor necrosis factor (TNF) therapy with TNF inhibitors (TNFi), etanercept, infliximab or adalimumab. In the present study, 11 patients who were TNFi drug naive were started on TNFi at a time of high disease activity. Of these, all cases were positive for rheumatoid factor and 9 cases tested were positive for anti-citrullinated peptide (anti-CCP) antibodies prior to TNFi treatment. Peripheral blood mononuclear cells (PBMCs) and serum were collected from all patients before and after TNFi therapy. Serum was assayed for ANAs over time. Total cellular RNA was extracted from PBMCs and assessed using Illumina arrays. Gene expression profiles were examined for alterations in key effector pathways. After 3 or more months on TNFi, 6 patients converted to ANA-positivity. Analysis of transcripts from patients with RA who converted to ANA-positivity after 3 months on TNFi identified complex gene expression profiles that reflected a reduction in cell adhesion, cell stress and lipid metabolism transcripts. In summary, unique transcriptional profiles in PBMCs from patients with RA were observed after TNFi therapy. This pilot study suggests that transcriptional profiling is a precise method of measuring the impact of TNFi therapies and reveals novel pathways that likely influence the immune response.

19.
J Mol Med (Berl) ; 95(7): 753-765, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28357476

RESUMEN

Interferon alpha and its surrogates, including IP-10 and SIGLEC1, paralleled changes of disease activity in systemic lupus erythematosus (SLE). However, the whole blood interferon signature (WBIFNS)-the current standard for type I IFN assessment in SLE-does not correlate with SLE disease activity in individual patients over time. The underlying causes for this apparent contradiction have not been convincingly demonstrated. Using a multicenter dataset of gene expression data from leukocyte subsets in SLE, we identify distinctive subset-specific contributions to the WBIFNS. In a subsequent analysis, the effects of type I interferon on cellular blood composition in patients with SLE and hepatitis B were also studied over time. We found that type I interferon mediates significant alterations in whole blood composition, including a neutropenia and relative lymphocytosis. Given different effects of type 1 interferon on different leukocyte subsets, these shifts confound measurement of a type 1 interferon signature in whole blood. To minimize and overcome these limitations of the WBIFNS, we suggest to measure IFN-induced transcripts or proteins in a specific leukocyte subset to improve clinical impact of interferon biomarkers. KEY MESSAGES: Myeloid cells contribute more to the WBIFNS in SLE than their lymphocytic counterpart. Very similar leukocyte subsets reveal distinctive IFN signatures. IFN alpha mixes up composition of blood and leads to a preferential neutropenia, yielding relative lymphocytosis.


Asunto(s)
Hepatitis B/genética , Hepatitis C/genética , Interferón Tipo I/genética , Leucocitos/patología , Lupus Eritematoso Sistémico/genética , Transcriptoma , Adulto , Biomarcadores/análisis , Biomarcadores/sangre , Femenino , Hepatitis B/sangre , Hepatitis B/patología , Hepatitis C/sangre , Hepatitis C/patología , Humanos , Interferón Tipo I/análisis , Interferón Tipo I/sangre , Leucocitos/metabolismo , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/patología , Linfocitosis/sangre , Linfocitosis/genética , Linfocitosis/patología , Masculino , Persona de Mediana Edad
20.
Inflammation ; 39(3): 1169-76, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27083877

RESUMEN

CD354, Triggering Receptor of Myeloid Cells-1 (TREM-1), is a potent amplifier of myeloid immune responses. Our goal was to determine the expression and function of TREM-1 in immune-mediated nephritis. An anti-glomerular basement membrane antibody (anti-GBM)-induced nephritis model was employed, where mice were sensitized with rabbit IgG followed by anti-GBM serum to induce disease. Anti-GBM-treated 129x1/svJ mice developed severe nephritis whereas C57BL/6 (B6) mice were resistant to disease. Anti-GBM disease resulted in elevated renal TREM-1 messenger RNA (mRNA) and protein levels and increased urine TREM-1 levels in 129x1/svJ. TREM-1 blockade with an inhibitory peptide, LP17, inhibited proteinuria and renal disease as measured by glomerulonephritis class, severity of tubulointerstitial disease, crescent formation, and inflammatory cell infiltrates. In sum, TREM-1 is upregulated in renal inflammation and plays a vital role in driving disease. Thus, TREM-1 blockade emerges as a potential therapeutic avenue for immune-mediated renal diseases such as lupus nephritis.


Asunto(s)
Nefritis Lúpica/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/análisis , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/análisis , Animales , Autoanticuerpos/farmacología , Modelos Animales de Enfermedad , Nefritis Lúpica/inducido químicamente , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos , ARN Mensajero/biosíntesis , Conejos , Receptores Inmunológicos/genética , Receptor Activador Expresado en Células Mieloides 1 , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA