Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Trials ; 24(1): 180, 2023 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-36906660

RESUMEN

BACKGROUND: Sequential B cell-targeted immunotherapy with BAFF antagonism (belimumab) and B cell depletion (rituximab) may enhance B cell targeting in ANCA-associated vasculitis (AAV) through several mechanisms. METHODS: Study design: COMBIVAS is a randomised, double-blind, placebo-controlled trial designed to assess the mechanistic effects of sequential therapy of belimumab and rituximab in patients with active PR3 AAV. The recruitment target is 30 patients who meet the criteria for inclusion in the per-protocol analysis. Thirty-six participants have been randomised to one of the two treatment groups in a 1:1 ratio: either rituximab plus belimumab or rituximab plus placebo (both groups with the same tapering corticosteroid regimen), and recruitment is now closed (final patient enrolled April 2021). For each patient, the trial will last for 2 years comprising a 12-month treatment period followed by a 12-month follow-up period. PARTICIPANTS: Participants have been recruited from five of seven UK trial sites. Eligibility criteria were age ≥ 18 years and a diagnosis of AAV with active disease (newly diagnosed or relapsing disease), along with a concurrent positive test for PR3 ANCA by ELISA. INTERVENTIONS: Rituximab 1000 mg was administered by intravenous infusions on day 8 and day 22. Weekly subcutaneous injections of 200 mg belimumab or placebo were initiated a week before rituximab on day 1 and then weekly through to week 51. All participants received a relatively low prednisolone (20 mg/day) starting dose from day 1 followed by a protocol-specified corticosteroid taper aiming for complete cessation by 3 months. OUTCOMES: The primary endpoint of this study is time to PR3 ANCA negativity. Key secondary outcomes include change from baseline in naïve, transitional, memory, plasmablast B cell subsets (by flow cytometry) in the blood at months 3, 12, 18 and 24; time to clinical remission; time to relapse; and incidence of serious adverse events. Exploratory biomarker assessments include assessment of B cell receptor clonality, B cell and T cell functional assays, whole blood transcriptomic analysis and urinary lymphocyte and proteomic analysis. Inguinal lymph node and nasal mucosal biopsies have been performed on a subgroup of patients at baseline and month 3. DISCUSSION: This experimental medicine study provides a unique opportunity to gain detailed insights into the immunological mechanisms of belimumab-rituximab sequential therapy across multiple body compartments in the setting of AAV. TRIAL REGISTRATION: ClinicalTrials.gov NCT03967925. Registered on May 30, 2019.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos , Inmunosupresores , Humanos , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/diagnóstico , Anticuerpos Anticitoplasma de Neutrófilos/uso terapéutico , Inmunosupresores/efectos adversos , Proteómica , Ensayos Clínicos Controlados Aleatorios como Asunto , Rituximab , Resultado del Tratamiento
2.
JCI Insight ; 7(23)2022 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-36477362

RESUMEN

BACKGROUNDPrimary Sjögren's syndrome (pSS) is characterized by B cell hyperactivity and elevated B-lymphocyte stimulator (BLyS). Anti-BLyS treatment (e.g., belimumab) increases peripheral memory B cells; decreases naive, activated, and plasma B cell subsets; and increases stringency on B cell selection during reconstitution. Anti-CD20 therapeutics (e.g., rituximab) bind and deplete CD20-expressing B cells in circulation but are less effective in depleting tissue-resident CD20+ B cells. Combined, these 2 mechanisms may achieve synergistic effects.METHODSThis 68-week, phase II, double-blind study (GSK study 201842) randomized 86 adult patients with active pSS to 1 of 4 arms: placebo, s.c. belimumab, i.v. rituximab, or sequential belimumab + rituximab.RESULTSOverall, 60 patients completed treatment and follow-up until week 68. The incidence of adverse events (AEs) and drug-related AEs was similar across groups. Infections/infestations were the most common AEs, and no serious infections of special interest occurred. Near-complete depletion of minor salivary gland CD20+ B cells and a greater and more sustained depletion of peripheral CD19+ B cells were observed with belimumab + rituximab versus monotherapies. With belimumab + rituximab, reconstitution of peripheral B cells occurred, but it was delayed compared with rituximab. At week 68, mean (± standard error) total EULAR Sjögren's syndrome disease activity index scores decreased from 11.0 (1.17) at baseline to 5.0 (1.27) for belimumab + rituximab and 10.4 (1.36) to 8.6 (1.57) for placebo.CONCLUSIONThe safety profile of belimumab + rituximab in pSS was consistent with the monotherapies. Belimumab + rituximab induced enhanced salivary gland B cell depletion relative to the monotherapies, potentially leading to improved clinical outcomes.TRIAL REGISTRATIONClinicalTrials.gov NCT02631538.FUNDINGFunding was provided by GSK.


Asunto(s)
Síndrome de Sjögren , Humanos , Rituximab/uso terapéutico , Síndrome de Sjögren/tratamiento farmacológico
3.
Ultrasound Med Biol ; 48(6): 1019-1032, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35307235

RESUMEN

Ultrasound has previously been demonstrated to non-invasively cause tissue disruption. Small animal studies have demonstrated that this effect can be enhanced by contrast microbubbles and has the potential to be clinically beneficial in techniques such as targeted drug delivery or enhancing liquid biopsies when a physical biopsy may be inappropriate. Cavitating microbubbles in close proximity to cells increases membrane permeability, allowing small intracellular molecules to leak into the extracellular space. This study sought to establish whether cavitating microbubbles could liberate cell-specific miRNAs, augmenting biomarker detection for non-invasive liquid biopsies. Insonating human polarized renal proximal tubular epithelial cells (RPTECs), in the presence of SonoVue microbubbles, revealed that cellular health could be maintained while achieving the release of miRNAs, miR-21, miR-30e, miR-192 and miR-194 (respectively, 10.9-fold, 7.17-fold, 5.95-fold and 5.36-fold). To examine the mechanism of release, RPTECs expressing enhanced green fluorescent protein were generated and the protein successfully liberated. Cell polarization, cellular phenotype and cell viability after sonoporation were measured by a number of techniques. Ultrastructural studies using electron microscopy showed gap-junction disruption and pore formation on cellular surfaces. These studies revealed that cell-specific miRNAs can be non-specifically liberated from RPTECs by sonoporation without a significant decrease in cell viability.


Asunto(s)
MicroARNs , Animales , Biomarcadores , Permeabilidad de la Membrana Celular , Células Epiteliales , Humanos , Microburbujas
4.
JCI Insight ; 5(24)2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33328386

RESUMEN

Small noncoding RNAs, miRNAs (miRNAs), are emerging as important modulators in the pathogenesis of kidney disease, with potential as biomarkers of kidney disease onset, progression, or therapeutic efficacy. Bulk tissue small RNA-sequencing (sRNA-Seq) and microarrays are widely used to identify dysregulated miRNA expression but are limited by the lack of precision regarding the cellular origin of the miRNA. In this study, we performed cell-specific sRNA-Seq on tubular cells, endothelial cells, PDGFR-ß+ cells, and macrophages isolated from injured and repairing kidneys in the murine reversible unilateral ureteric obstruction model. We devised an unbiased bioinformatics pipeline to define the miRNA enrichment within these cell populations, constructing a miRNA catalog of injury and repair. Our analysis revealed that a significant proportion of cell-specific miRNAs in healthy animals were no longer specific following injury. We then applied this knowledge of the relative cell specificity of miRNAs to deconvolute bulk miRNA expression profiles in the renal cortex in murine models and human kidney disease. Finally, we used our data-driven approach to rationally select macrophage-enriched miR-16-5p and miR-18a-5p and demonstrate that they are promising urinary biomarkers of acute kidney injury in renal transplant recipients.


Asunto(s)
Lesión Renal Aguda/genética , MicroARNs/genética , Especificidad de Órganos/genética , Animales , Biomarcadores , Biología Computacional/métodos , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Riñón/metabolismo , Túbulos Renales/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo
5.
Arthritis Res Ther ; 22(1): 102, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366280

RESUMEN

BACKGROUND: Elevated B lymphocyte stimulator (BLyS) levels in patients with systemic lupus erythematosus (SLE) correlate positively with disease activity; BLyS expression is directly linked to interferon (IFN) pathway activation. This post hoc meta-analysis of BLISS-52 and BLISS-76 explored the relationship between baseline BLyS mRNA/protein levels and/or type 1 IFN-inducible gene signature (IFN-1) and responses to the BLyS-targeting monoclonal antibody belimumab in SLE. METHODS: In BLISS-52 and BLISS-76, patients with autoantibody-positive SLE and a SELENA-SLEDAI score ≥ 6 and receiving stable standard SLE therapy were randomised to intravenous belimumab 10 mg/kg or placebo, plus standard of care (SoC), for 52 or 76 weeks. For this post hoc meta-analysis, patients with an appropriate mRNA sample were stratified by BLyS mRNA expression (tertiles: high/medium/low; revised quantiles: high/low), IFN-1 mRNA expression (high/low) and BLyS protein level (high/low). Co-primary endpoints were correlation between baseline BLyS and IFN-1 mRNA levels and SLE Responder Index (SRI)4 response at week 52 within BLyS/IFN-1 subgroups. Secondary endpoints included time to first severe SELENA-SLEDAI Flare Index (SFI) flare. RESULTS: Of 554 patients included in this analysis, 281 had received belimumab and 273 had received placebo. Baseline BLyS and IFN-1 mRNA levels were highly correlated (Spearman's rank correlation coefficient 0.7799; 95% confidence interval [CI] 0.7451, 0.8106; p < 0.0001). The proportion of SRI4 responders was higher with belimumab versus placebo in all subgroups, but the difference reached statistical significance in the medium BLyS mRNA tertile (odds ratio [OR] 2.17; 95% CI 1.16, 4.04; p = 0.0153), high BLyS mRNA quantile (OR 1.58; 95% CI 1.02, 2.44; p = 0.0402), high IFN-1 mRNA (OR 1.58; 95% CI: 1.08, 2.31; p = 0.0186) and high BLyS protein (OR 3.57; 95% CI 1.63, 7.83; p = 0.0015) subgroups only. The risk of severe SFI flare was significantly lower with belimumab than placebo in the high BLyS mRNA quantile (hazard ratio [HR] 0.59; 95% CI 0.36, 0.97; p = 0.0371) and high BLyS protein (HR 0.39; 95% CI 0.19, 0.79; p = 0.0090) subgroups. CONCLUSIONS: This post hoc meta-analysis demonstrated a tendency towards improved response to add-on intravenous belimumab 10 mg/kg versus SoC alone in patients with high baseline BLyS protein and IFN-1 mRNA levels and medium/high BLyS mRNA levels.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Activador de Células B , Interferón Tipo I/genética , Lupus Eritematoso Sistémico , Factor Activador de Células B/genética , Femenino , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Masculino , ARN Mensajero , Ensayos Clínicos Controlados Aleatorios como Asunto , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
6.
Nephrol Dial Transplant ; 35(4): 599-606, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31243451

RESUMEN

BACKGROUND: Immunosuppressant drugs reduce proteinuria and anti-phospholipase A2 receptor autoantibodies (PLA2R-Ab) in primary membranous nephropathy (PMN) with varying success and associated toxicities. This study aimed to evaluate the effect of belimumab on proteinuria and PLA2R-Ab in participants with PMN. METHODS: In this prospective, open-label, experimental medicine study, 14 participants with PMN and persistent nephrotic-range proteinuria received up to 2 years belimumab monotherapy (10 mg/kg, every 4 weeks). Changes in proteinuria (urinary protein:creatinine ratio), PLA2R-Ab, albumin, cholesterol, B-cell subsets and pharmacokinetics were analysed during treatment and up to 6 months after treatment. RESULTS: Eleven participants completed to the primary endpoint (Week 28) and nine participants completed the study. In the intention-to-treat population population, baseline proteinuria of 724 mg/mmol [95% confidence interval (CI) 579-906] decreased to 498 mg/mmol (95% CI 383-649) and 130 mg/mmol (95% CI 54-312) at Weeks 28 and 104, respectively, with changes statistically significant from Week 36 (n = 11, P = 0.047). PLA2R-Ab decreased from 174 RU/mL (95% CI 79-384) at baseline to 46 RU/mL (95% CI 16-132) and 4 RU/mL (95% CI 2-6) at Weeks 28 and 104, respectively, becoming statistically significant by Week 12 (n = 13, P = 0.02). Nine participants achieved partial (n = 8) or complete (n = 1) remission. Participants with abnormal albumin and/or cholesterol at baseline gained normal/near normal levels by the last follow-up. Adverse events were consistent with those expected in this population. CONCLUSIONS: Belimumab treatment in participants with PMN can reduce PLA2R-Ab and subsequently proteinuria, important preludes to remission induction.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Autoanticuerpos/inmunología , Glomerulonefritis Membranosa/complicaciones , Inmunosupresores/uso terapéutico , Proteinuria/tratamiento farmacológico , Receptores de Fosfolipasa A2/inmunología , Adulto , Anciano , Autoanticuerpos/efectos de los fármacos , Femenino , Glomerulonefritis Membranosa/inmunología , Glomerulonefritis Membranosa/patología , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Proteinuria/etiología , Proteinuria/patología , Inducción de Remisión , Adulto Joven
7.
BMJ Open ; 9(3): e025687, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30898822

RESUMEN

INTRODUCTION: Belimumab, an anti-B-lymphocyte-stimulator antibody, is approved for the treatment of active, autoantibody-positive systemic lupus erythematosus (SLE). Rituximab, a B cell-depleting anti-CD20 antibody, remains in the SLE treatment armamentarium despite failed trials in lupus nephritis and extrarenal lupus. These biologics, which operate through complementary mechanisms, might result in an enhanced depletion of circulating and tissue-resident autoreactive B lymphocytes when administered together. Thus, belimumab and rituximab combination may be a highly effective treatment of SLE. This study aims to evaluate and compare the efficacy, safety and tolerability of subcutaneous (SC) belimumab and a single cycle of rituximab in patients with SLE with belimumab alone. METHODS AND ANALYSIS: BLISS-BELIEVE is a three-arm, randomised, double-blind, placebo-controlled, 104-week superiority study. Two hundred adults with SLE will be randomised 1:2:1 to arm A, belimumab SC 200 mg/week for 52 weeks plus placebo at weeks 4 and 6; arm B, belimumab SC 200 mg/week for 52 weeks plus rituximab 1000 mg at weeks 4 and 6; arm C, belimumab SC 200 mg/week plus standard of care for 104 weeks. The 52-week treatment period (arms A and B) is followed by a 52-week observational phase. The primary efficacy endpoint is the proportion of patients with disease control (SLE Disease Activity Index (SLEDAI)-2K≤2, without immunosuppressants and with a prednisone-equivalent dose of ≤5 mg/day) at week 52. Major secondary efficacy endpoints are the proportion of patients in clinical remission (defined as SLEDAI-2K=0, without immunosuppressants and corticosteroids) at week 64, and the proportion of patients with disease control at week 104. Safety endpoints include the incidence of adverse events (AEs), serious AEs and AEs of special interest. ETHICS AND DISSEMINATION: Within 6 months of the study's primary manuscript publication, anonymised individual participant data and study documents can be requested for further research from www.clinicalstudydatarequest.com. TRIAL REGISTRATION NUMBER: NCT03312907; Pre-results.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Inmunosupresores/administración & dosificación , Lupus Eritematoso Sistémico/tratamiento farmacológico , Rituximab/administración & dosificación , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Ensayos Clínicos Fase III como Asunto , Método Doble Ciego , Femenino , Humanos , Inmunosupresores/efectos adversos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Estudios Multicéntricos como Asunto , Prednisona/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto , Rituximab/efectos adversos , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
9.
Arthritis Rheumatol ; 71(6): 952-963, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30666823

RESUMEN

OBJECTIVE: To evaluate the safety and efficacy of belimumab as adjunctive therapy to maintain remission in antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). METHODS: In this multicenter, double-blind, placebo-controlled study, patients with AAV (ages ≥18 years) were randomized 1:1 to receive azathioprine (2 mg/kg/day), low-dose oral glucocorticoids (≤10 mg/day), and either intravenous belimumab (10 mg/kg) or placebo, following remission induction with rituximab or cyclophosphamide along with glucocorticoids. The primary end point was time to first protocol-specified event (PSE), with first PSE defined as a Birmingham Vasculitis Activity Score (BVAS) of ≥6, presence of ≥1 major BVAS item, or receipt of prohibited medications for any reason, resulting in treatment failure (adjusted for ANCA type [proteinase 3 (PR3) or myeloperoxidase (MPO)], disease stage at induction, and induction regimen). Vasculitis relapse was defined as the PSE of either a BVAS activity score of ≥6 or receipt of prohibited medications for vasculitis. Changes in treatment practice led to truncation of the study population from ~300 patients to ~100 patients. RESULTS: The intent-to-treat population totaled 105 patients with AAV, of whom 52 (40 with PR3-ANCAs, 12 with MPO-ANCAs) received placebo and 53 (41 with PR3-ANCAs, 12 with MPO-ANCAs) received belimumab; 27 of the patients were in rituximab-induced disease remission, while 78 were in cyclophosphamide-induced disease remission at baseline. Compared with placebo, treatment with belimumab did not reduce the risk of a PSE (adjusted hazard ratio [HR] 1.07, 95% confidence interval [95% CI] 0.44-2.59; P = 0.884) or vasculitis relapse (adjusted HR 0.88, 95% CI 0.29-2.65; P = 0.821). The overall rate of PSEs was low (11 [21.2%] of 52 patients receiving placebo, 10 [18.9%] of 53 patients receiving belimumab). Vasculitis relapse in the placebo group (n = 8) occurred independent of the induction regimen, disease stage, or ANCA type. All vasculitis relapses in the belimumab group (n = 6) occurred in patients who had PR3-ANCA-associated vasculitis with cyclophosphamide-induced disease remission. Adverse events occurred in 49 (92.5%) of 53 patients receiving belimumab and 43 (82.7%) of 52 patients receiving placebo, with no new safety concerns. CONCLUSION: Belimumab plus azathioprine and glucocorticoids for the maintenance of remission in AAV did not reduce the risk of relapse.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Azatioprina/uso terapéutico , Inmunosupresores/uso terapéutico , Adulto , Anciano , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Linfocitos B/citología , Ciclofosfamida/uso terapéutico , Método Doble Ciego , Quimioterapia Combinada , Femenino , Glucocorticoides/uso terapéutico , Humanos , Inmunoglobulinas/inmunología , Infecciones/epidemiología , Quimioterapia de Mantención , Masculino , Persona de Mediana Edad , Inducción de Remisión , Rituximab/uso terapéutico , Resultado del Tratamiento
10.
Lancet ; 391(10140): 2619-2630, 2018 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-29910042

RESUMEN

BACKGROUND: B cells produce alloantibodies and activate alloreactive T cells, negatively affecting kidney transplant survival. By contrast, regulatory B cells are associated with transplant tolerance. Immunotherapies are needed that inhibit B-cell effector function, including antibody secretion, while sparing regulators and minimising infection risk. B lymphocyte stimulator (BLyS) is a cytokine that promotes B-cell activation and has not previously been targeted in kidney transplant recipients. We aimed to determine the safety and activity of an anti-BLyS antibody, belimumab, in addition to standard-of-care immunosuppression in adult kidney transplant recipients. We used an experimental medicine study design with multiple secondary and exploratory endpoints to gain further insight into the effect of belimumab on the generation of de-novo IgG and on the regulatory B-cell compartment. METHODS: We undertook a double-blind, randomised, placebo-controlled phase 2 trial of belimumab, in addition to standard-of-care immunosuppression (basiliximab, mycophenolate mofetil, tacrolimus, and prednisolone) at two centres, Addenbrooke's Hospital, Cambridge, UK, and Guy's and St Thomas' Hospital, London, UK. Participants were eligible if they were aged 18-75 years and receiving a kidney transplant and were planned to receive standard-of-care immunosuppression. Participants were randomly assigned (1:1) to receive either intravenous belimumab 10 mg per kg bodyweight or placebo, given at day 0, 14, and 28, and then every 4 weeks for a total of seven infusions. The co-primary endpoints were safety and change in the concentration of naive B cells from baseline to week 24, both of which were analysed in all patients who received a transplant and at least one dose of drug or placebo (the modified intention-to-treat [mITT] population). This trial has been completed and is registered with ClinicalTrials.gov, NCT01536379, and EudraCT, 2011-006215-56. FINDINGS: Between Sept 13, 2013, and Feb 8, 2015, of 303 patients assessed for eligibility, 28 kidney transplant recipients were randomly assigned to receive belimumab (n=14) or placebo (n=14). 25 patients (12 [86%] patients assigned to the belimumab group and 13 [93%] patients assigned to the placebo group) received a transplant and were included in the mITT population. We observed similar proportions of adverse events in the belimumab and placebo groups, including serious infections (one [8%] of 12 in the belimumab group and five [38%] of 13 in the placebo group during the 6-month on-treatment phase; and none in the belimumab group and two [15%] in the placebo group during the 6-month follow-up). In the on-treatment phase, one patient in the placebo group died because of fatal myocardial infarction and acute cardiac failure. The co-primary endpoint of a reduction in naive B cells from baseline to week 24 was not met. Treatment with belimumab did not significantly reduce the number of naive B cells from baseline to week 24 (adjusted mean difference between the belimumab and placebo treatment groups -34·4 cells per µL, 95% CI -109·5 to 40·7). INTERPRETATION: Belimumab might be a useful adjunct to standard-of-care immunosuppression in renal transplantation, with no major increased risk of infection and potential beneficial effects on humoral alloimmunity. FUNDING: GlaxoSmithKline.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Supervivencia de Injerto/efectos de los fármacos , Terapia de Inmunosupresión/métodos , Inmunosupresores/administración & dosificación , Trasplante de Riñón/métodos , Administración Intravenosa , Adulto , Anciano , Método Doble Ciego , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad
11.
Haematologica ; 101(6): 698-706, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26969086

RESUMEN

Primary immune thrombocytopenia is an autoimmune disorder in which platelet destruction is a consequence of both B- and T-cell dysregulation. Flow cytometry was used to further characterize the B- and T-cell compartments in a cross-sectional cohort of 26 immune thrombocytopenia patients including antiplatelet antibody positive (n=14) and negative (n=12) patients exposed to a range of therapies, and a cohort of matched healthy volunteers. Markers for B-cell activating factor and its receptors, relevant B-cell activation markers (CD95 and CD21) and markers for CD4(+) T-cell subsets, including circulating T-follicular helper-like cells, were included. Our results indicate that an expanded population of CD95(+) naïve B cells correlated with disease activity in immune thrombocytopenia patients regardless of treatment status. A population of CD21-naïve B cells was specifically expanded in autoantibody-positive immune thrombocytopenia patients. Furthermore, the B-cell maturation antigen, a receptor for B-cell activating factor, was consistently and strongly up-regulated on plasmablasts from immune thrombocytopenia patients. These observations have parallels in other autoantibody-mediated diseases and suggest that loss of peripheral tolerance in naïve B cells may be an important component of immune thrombocytopenia pathogenesis. Moreover, the B-cell maturation antigen represents a potential target for plasma cell directed therapies in immune thrombocytopenia.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Fenotipo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/metabolismo , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Factor Activador de Células B/sangre , Factor Activador de Células B/metabolismo , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Biomarcadores , Plaquetas/inmunología , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Células Plasmáticas/patología , Púrpura Trombocitopénica Idiopática/diagnóstico , Púrpura Trombocitopénica Idiopática/terapia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
12.
Blood ; 116(23): 4978-89, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-20813901

RESUMEN

The generation of reactive oxygen species (ROS) by the nicotinamide adenine dinucleotide phosphate oxidase is an important mechanism by which neutrophils kill pathogens. The oxidase is composed of a membrane-bound cytochrome and 4 soluble proteins (p67(phox), p40(phox), p47(phox), and GTP-Rac). These components form an active complex at the correct time and subcellular location through a series of incompletely understood mutual interactions, regulated, in part, by GTP/GDP exchange on Rac, protein phosphorylation, and binding to lipid messengers. We have used a variety of assays to follow the spatiotemporal assembly of the oxidase in genetically engineered primary mouse neutrophils, during phagocytosis of both serum- and immunoglobulin G-opsonized targets. The oxidase assembles directly on serum-Staphylococcus aureus-containing phagosomes within seconds of phagosome formation; this process is only partially dependent (∼ 30%) on PtdIns3P binding to p40(phox), but totally dependent on Rac1/2 binding to p67(phox). In contrast, in response to immunoglobulin G-targets, the oxidase first assembles on a tubulovesicular compartment that develops at sites of granule fusion to the base of the emerging phagosome; oxidase assembly and activation is highly dependent on both PtdIns3P-p40(phox) and Rac2-p67(phox) interactions and delivery to the phagosome is regulated by Rab27a. These results define a novel pathway for oxidase assembly downstream of FcR-activation.


Asunto(s)
NADPH Oxidasas/metabolismo , Neutrófilos/enzimología , Fagocitosis/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Activación Enzimática/fisiología , Humanos , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Fosfatos de Fosfatidilinositol/inmunología , Fosfoproteínas/inmunología , Fosfoproteínas/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores Fc/inmunología , Proteínas de Unión al GTP rac/inmunología
13.
J Exp Med ; 207(4): 837-53, 2010 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-20308364

RESUMEN

Rac1 and Rac2 GTPases transduce signals from multiple receptors leading to cell migration, adhesion, proliferation, and survival. In the absence of Rac1 and Rac2, B cell development is arrested at an IgD- transitional B cell stage that we term transitional type 0 (T0). We show that T0 cells cannot enter the white pulp of the spleen until they mature into the T1 and T2 stages, and that this entry into the white pulp requires integrin and chemokine receptor signaling and is required for cell survival. In the absence of Rac1 and Rac2, transitional B cells are unable to migrate in response to chemokines and cannot enter the splenic white pulp. We propose that loss of Rac1 and Rac2 causes arrest at the T0 stage at least in part because transitional B cells need to migrate into the white pulp to receive survival signals. Finally, we show that in the absence of Syk, a kinase that transduces B cell antigen receptor signals required for positive selection, development is arrested at the same T0 stage, with transitional B cells excluded from the white pulp. Thus, these studies identify a novel developmental checkpoint that coincides with B cell positive selection.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular , Movimiento Celular , Bazo/citología , Proteínas de Unión al GTP rac/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Antígenos CD/metabolismo , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Adhesión Celular/genética , Adhesión Celular/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Supervivencia Celular/genética , Quimiocinas/farmacología , Inmunoglobulina D/metabolismo , Integrinas/antagonistas & inhibidores , Integrinas/inmunología , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Neuropéptidos/genética , Toxina del Pertussis/farmacología , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-vav/genética , Receptores CXCR4/antagonistas & inhibidores , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Bazo/inmunología , Quinasa Syk , Proteína bcl-X/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1 , Proteínas de Unión al GTP rap1/metabolismo , Proteína RCA2 de Unión a GTP
14.
Nat Rev Immunol ; 9(9): 630-44, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19696767

RESUMEN

Rho family GTPases, and the proteins that regulate them, have important roles in many cellular processes, including cell division, survival, migration and adhesion. Although most of our understanding of these proteins has come from studies using cell lines, more recent gene targeting studies in mice are providing insights into the in vivo function of these proteins. Here we review recent progress revealing crucial roles for these proteins in lymphocyte development, activation, differentiation and migration. The emerging picture shows that Rho family GTPases transduce signals from receptors for antigens, chemokines and cytokines, as well as adhesion molecules and pattern recognition receptors, and that they function as focal points for crosstalk between different signalling pathways.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T/inmunología , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/clasificación , Animales , Linfocitos B/enzimología , Movimiento Celular/inmunología , Citoesqueleto/inmunología , Citoesqueleto/metabolismo , Humanos , Integrinas/inmunología , Integrinas/metabolismo , Ratones , Filogenia , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/inmunología , Linfocitos T/enzimología
15.
J Immunol ; 180(6): 4308-15, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18322244

RESUMEN

Neutrophils are the first immune cells to migrate into infected tissue sites. Therefore an important step in the initiation of an immune response is the synthesis of the neutrophil-recruiting chemokines. In this in vivo study in mice, we show that resident tissue macrophages are the source of the major neutrophil chemoattractants, KC and MIP-2. Synthesis of these chemokines is rapidly regulated at the transcriptional level by signaling through TLR2, TLR3, and TLR4 that have diverse specificities for pathogens. The major and alternative TLR signaling pathways are characterized by the adaptor proteins MyD88 or TRIF, respectively. KC and MIP-2 are both produced by signaling through MyD88. However MIP-2, but not KC, is also synthesized through the TRIF adaptor protein, identifying it as a new product of this alternative pathway. Use of both pathways by TLR4 ensures maximal levels of KC and MIP-2 that lead to robust neutrophil recruitment. However the MIP-2 generated exclusively by the TRIF pathway is still sufficient to cause an influx of neutrophils. In summary we show that TLR signaling by tissue macrophages directly controls the synthesis of neutrophil-attracting chemokines that are essential for the earliest recruitment step in the innate immune response to microbial challenge.


Asunto(s)
Quimiocina CXCL1/biosíntesis , Quimiocina CXCL2/biosíntesis , Macrófagos/inmunología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/fisiología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Quimiocina CXCL1/genética , Quimiocina CXCL2/genética , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/metabolismo
16.
J Cell Sci ; 119(Pt 13): 2749-57, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16772332

RESUMEN

Rac GTPases are believed to contribute to migration in leukocytes by transducing signals from cell surface receptors to the actin and microtubule cytoskeletons. Mammals have three closely related Rac isoforms, Rac1, Rac2 and Rac3, and it is widely assumed that cell migration requires the activity of these Rac GTPases. We have previously shown that Rac1-null mouse macrophages have altered cell shape and reduced membrane ruffling but normal migration speed. Here we investigate the behaviour of macrophages lacking Rac2 (Rac2(-/-)) or Rac1 and Rac2 (Rac1/2(-/-)). Rac2(-/-) macrophages have reduced F-actin levels and lack podosomes, which are integrin-based adhesion sites, and their migration speed is similar to or slightly slower than wild-type macrophages, depending on the substrate. Unexpectedly, Rac1/2(-/-) macrophages, which do not express Rac1, Rac2 or Rac3, migrate at a similar speed to wild-type macrophages on a variety of substrates and perform chemotaxis normally, although their morphology and mode of migration is altered. However, Rac1(-/-) and Rac1/2(-/-) but not Rac2(-/-) macrophages are impaired in their ability to invade through Matrigel. Together, these data show that Rac1 and Rac2 have distinct roles in regulating cell morphology, migration and invasion, but are not essential for macrophage migration or chemotaxis.


Asunto(s)
Movimiento Celular/fisiología , Macrófagos/fisiología , Morfogénesis/fisiología , Neuropéptidos/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Células de la Médula Ósea/fisiología , Células Cultivadas , Quimiotaxis/genética , Colágeno/metabolismo , Combinación de Medicamentos , Fibronectinas/farmacología , Laminina/metabolismo , Laminina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/deficiencia , Proteoglicanos/metabolismo , Seudópodos/metabolismo , Seudópodos/fisiología , Eliminación de Secuencia , Proteínas de Unión al GTP rac/deficiencia , Proteína de Unión al GTP rac1 , Proteína RCA2 de Unión a GTP
17.
J Biol Chem ; 280(47): 39474-84, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16195235

RESUMEN

The role of Rac family proteins in platelet spreading on matrix proteins under static and flow conditions has been investigated by using Rac-deficient platelets. Murine platelets form filopodia and undergo limited spreading on fibrinogen independent of Rac1 and Rac2. In the presence of thrombin, marked lamellipodia formation is observed on fibrinogen, which is abrogated in the absence of Rac1. However, Rac1 is not required for thrombin-induced aggregation or elevation of F-actin levels. Formation of lamellipodia on collagen and laminin is also Rac1-dependent. Analysis of platelet adhesion dynamics on collagen under flow conditions in vitro revealed that Rac1 is required for platelet aggregate stability at arterial rates of shear, as evidenced by a dramatic increase in platelet embolization. Furthermore, studies employing intravital microscopy demonstrated that Rac1 plays a critical role in the development of stable thrombi at sites of vascular injury in vivo. Thus, our data demonstrated that Rac1 is essential for lamellipodia formation in platelets and indicated that Rac1 is required for aggregate integrity leading to thrombus formation under physiologically relevant levels of shear both in vitro and in vivo.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/ultraestructura , Neuropéptidos/sangre , Seudópodos/metabolismo , Seudópodos/ultraestructura , Proteínas de Unión al GTP rac/sangre , Proteína de Unión al GTP rac1/sangre , Animales , Fibrinógeno , Hemorreología , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Neuropéptidos/deficiencia , Neuropéptidos/genética , Adhesividad Plaquetaria , Agregación Plaquetaria , Propiedades de Superficie , Proteínas de Unión al GTP rac/deficiencia , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
18.
Blood ; 102(1): 328-35, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12623845

RESUMEN

Early neutrophil entry into an inflammatory site is thought to mediate a chemokine switch, inducing subsequent monocyte recruitment through the regulation of monocyte chemoattractant protein-1 (MCP-1) release. As the murine monocyte is poorly characterized and difficult to identify, there has been little examination of either its early recruitment in inflammatory models or of the factors that influence its early migration. The phenotyping of rapidly recruited inflammatory leukocytes with 7/4 and Gr-1 monoclonal antibodies (mAbs) identifies 2 distinct populations, which we characterize as murine monocytes and neutrophils. Monocytes migrate in the first 2 hours of inflammation making use of alpha4beta1 but not of Mac-1 or lymphocyte function-associated antigen-1 (LFA-1) integrins. Early migration is dependent on MCP-1, but neither MCP-1 release nor monocyte recruitment is affected by the reduced neutrophil migration seen in LFA-1-/- mice. Endogenous peritoneal macrophages and mesothelial cells lining the peritoneum contain MCP-1, which is released following thioglycollate stimulation. The murine monocyte therefore responds rapidly to chemokines produced in situ by tissue cells at the site of inflammation with no requirement for prior influx of neutrophils.


Asunto(s)
Quimiotaxis de Leucocito , Monocitos/citología , Infiltración Neutrófila , Animales , Comunicación Celular , Quimiocina CCL2/análisis , Quimiocina CCL2/biosíntesis , Inflamación/inmunología , Inflamación/patología , Integrina alfa4/fisiología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Ratones , Ratones Noqueados , Peritonitis/inmunología , Peritonitis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...