Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(5): 607-619, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33833438

RESUMEN

FOXP3 deficiency in mice and in patients with immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome results in fatal autoimmunity by altering regulatory T (Treg) cells. CD4+ T cells in patients with IPEX syndrome and Foxp3-deficient mice were analyzed by single-cell cytometry and RNA-sequencing, revealing heterogeneous Treg-like cells, some very similar to normal Treg cells, others more distant. Conventional T cells showed no widespread activation or helper T cell bias, but a monomorphic disease signature affected all CD4+ T cells. This signature proved to be cell extrinsic since it was extinguished in mixed bone marrow chimeric mice and heterozygous mothers of patients with IPEX syndrome. Normal Treg cells exerted dominant suppression, quenching the disease signature and revealing in mutant Treg-like cells a small cluster of genes regulated cell-intrinsically by FOXP3, including key homeostatic regulators. We propose a two-step pathogenesis model: cell-intrinsic downregulation of core FOXP3-dependent genes destabilizes Treg cells, de-repressing systemic mediators that imprint the disease signature on all T cells, furthering Treg cell dysfunction. Accordingly, interleukin-2 treatment improved the Treg-like compartment and survival.


Asunto(s)
Diabetes Mellitus Tipo 1/congénito , Diarrea/genética , Factores de Transcripción Forkhead/deficiencia , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades del Sistema Inmune/congénito , Linfocitos T Reguladores/inmunología , Adolescente , Animales , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Conjuntos de Datos como Asunto , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diarrea/sangre , Diarrea/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Enfermedades Genéticas Ligadas al Cromosoma X/sangre , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Humanos , Enfermedades del Sistema Inmune/sangre , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/inmunología , Lactante , Masculino , Ratones , Ratones Transgénicos , Mutación , RNA-Seq , Análisis de la Célula Individual , Linfocitos T Reguladores/metabolismo , Adulto Joven
2.
Blood ; 137(17): 2326-2336, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33545713

RESUMEN

Immunodysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is caused by mutations in forkhead box P3 (FOXP3), which lead to the loss of function of regulatory T cells (Tregs) and the development of autoimmune manifestations early in life. The selective induction of a Treg program in autologous CD4+ T cells by FOXP3 gene transfer is a promising approach for curing IPEX. We have established a novel in vivo assay of Treg functionality, based on adoptive transfer of these cells into scurfy mice (an animal model of IPEX) and a combination of cyclophosphamide (Cy) conditioning and interleukin-2 (IL-2) treatment. This model highlighted the possibility of rescuing scurfy disease after the latter's onset. By using this in vivo model and an optimized lentiviral vector expressing human Foxp3 and, as a reporter, a truncated form of the low-affinity nerve growth factor receptor (ΔLNGFR), we demonstrated that the adoptive transfer of FOXP3-transduced scurfy CD4+ T cells enabled the long-term rescue of scurfy autoimmune disease. The efficiency was similar to that seen with wild-type Tregs. After in vivo expansion, the converted CD4FOXP3 cells recapitulated the transcriptomic core signature for Tregs. These findings demonstrate that FOXP3 expression converts CD4+ T cells into functional Tregs capable of controlling severe autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Linfocitos T CD4-Positivos/inmunología , Ciclofosfamida/farmacología , Factores de Transcripción Forkhead/genética , Enfermedades Genéticas Ligadas al Cromosoma X/prevención & control , Interleucina-2/farmacología , Linfocitos T Reguladores/inmunología , Animales , Antineoplásicos/farmacología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos
3.
Blood ; 135(15): 1219-1231, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32040546

RESUMEN

In gene therapy with human hematopoietic stem and progenitor cells (HSPCs), each gene-corrected cell and its progeny are marked in a unique way by the integrating vector. This feature enables lineages to be tracked by sampling blood cells and using DNA sequencing to identify the vector integration sites. Here, we studied 5 cell lineages (granulocytes, monocytes, T cells, B cells, and natural killer cells) in patients having undergone HSPC gene therapy for Wiskott-Aldrich syndrome or ß hemoglobinopathies. We found that the estimated minimum number of active, repopulating HSPCs (which ranged from 2000 to 50 000) was correlated with the number of HSPCs per kilogram infused. We sought to quantify the lineage output and dynamics of gene-modified clones; this is usually challenging because of sparse sampling of the various cell types during the analytical procedure, contamination during cell isolation, and different levels of vector marking in the various lineages. We therefore measured the residual contamination and corrected our statistical models accordingly to provide a rigorous analysis of the HSPC lineage output. A cluster analysis of the HSPC lineage output highlighted the existence of several stable, distinct differentiation programs, including myeloid-dominant, lymphoid-dominant, and balanced cell subsets. Our study evidenced the heterogeneous nature of the cell lineage output from HSPCs and provided methods for analyzing these complex data.


Asunto(s)
Células Clonales/citología , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Hemoglobinopatías/terapia , Síndrome de Wiskott-Aldrich/terapia , Diferenciación Celular , Rastreo Celular , Células Clonales/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Hemoglobinopatías/genética , Humanos , Síndrome de Wiskott-Aldrich/genética
4.
J Clin Apher ; 36(1): 12-19, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32854142

RESUMEN

BACKGROUND: Autologous and allogeneic hematopoietic stem cell transplantation of cytokine-mobilized peripheral blood stem cells (PBSCs) is increasingly used to treat patients with hematologic disorders. Different types of vascular access have been exploited for the apheresis procedure, including peripheral veins (PV) and central venous catheter (CVC). In some cases, PV access is unavailable. There are few published data on the efficiency and quality of harvesting with different types of vascular access. This study brings out complications and morbidity of this procedure linked to these different access. METHODS: We performed a comparative, retrospective, single-center study of hematopoietic stem cell collection using these two types of vascular access. We compared the efficiency and complication rate for 617 adults apheresis sessions in 401 patients and healthy donors, for PBSC collection via PV or CVC between 2010 and 2016. The quality of the HSC product was evaluated in terms of the total CD34 + count and neutrophil contamination. RESULTS: The PV and CVC groups did not differ significantly in terms of the quality of the apheresis product, mean ± SD CD34 + cells collected in PV group was 383.1 ± 402.7 × 10e6 and 298.8 ± 372.7 × 10e6 and the level of neutrophil contamination was 21.0 ± 17.8% in the PV group and 20.6 ± 18.4% in the CVC group. The complication rate did not differ between the two groups. CONCLUSION: The type of vascular access for apheresis hematopoietic stem cell harvesting must be determined by trained staff. Successful harvesting can be performed via PV then CVC is not needed or not available.


Asunto(s)
Eliminación de Componentes Sanguíneos/métodos , Cateterismo Venoso Central/métodos , Cateterismo Periférico/métodos , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/citología , Trasplante de Células Madre de Sangre Periférica/métodos , Adolescente , Adulto , Anciano , Cateterismo Venoso Central/efectos adversos , Cateterismo Periférico/efectos adversos , Femenino , Movilización de Célula Madre Hematopoyética/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
5.
J Clin Apher ; 36(6): 823-830, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34469617

RESUMEN

INTRODUCTION: Therapeutic plasma exchange (TPE) is acknowledged to be an effective treatment in life-threatening pediatric disorders. Apheresis for pediatric diseases has been poorly investigated, and most studies to date featured small numbers of patients and lacked control groups. The objective of the present study was to evaluate the tolerance of TPE in pediatric patients. MATERIALS AND METHODS: A retrospective cohort study via a web-based electronic case report form including pediatric patients referred for TPE between January 2005 and December 2014. RESULTS: A total of 78 patients (median [range] age: 9.8 [0.53-17.93]) and 731 TPE procedures were analyzed. The indications were antibody-mediated rejection (n = 33; 42%) and desensitization therapy (n = 5; 6%) after solid organ or hematopoietic stem cell transplantation, thrombotic microangiopathy (n = 17; 22%), pediatric inflammatory diseases (n = 16; 21%), kidney diseases (n = 6; 8%), and hyperviscosity syndrome (n = 1; 1%). On average, each patient underwent six procedures during the first session [range: 1-19]. In the 2 weeks following the start of a session, 72 patients (92%) presented a total of 311 adverse events (AEs) potentially related to TPE. The risk of AEs was not related to the indication for TPE, the intensity of care, venous access, plasma substitute use, or body weight. None of the deaths was related to the TPE. CONCLUSION: We studied one of the largest retrospective pediatric cohorts described to date. Our experience of TPE children's TPE feasibility concerned specific, life-threatening conditions and otherwise treatment-refractory diseases.


Asunto(s)
Cuidados Críticos/métodos , Intercambio Plasmático/métodos , Adolescente , Niño , Estudios de Factibilidad , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Inflamación/terapia , Unidades de Cuidado Intensivo Pediátrico , Enfermedades Renales/terapia , Masculino , Intercambio Plasmático/efectos adversos , Estudios Retrospectivos , Microangiopatías Trombóticas/terapia , Resultado del Tratamiento
6.
J Am Soc Nephrol ; 30(4): 692-709, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30850439

RESUMEN

BACKGROUND: Although anti-HLA antibodies (Abs) cause most antibody-mediated rejections of renal allografts, non-anti-HLA Abs have also been postulated to contribute. A better understanding of such Abs in rejection is needed. METHODS: We conducted a nationwide study to identify kidney transplant recipients without anti-HLA donor-specific Abs who experienced acute graft dysfunction within 3 months after transplantation and showed evidence of microvascular injury, called acute microvascular rejection (AMVR). We developed a crossmatch assay to assess serum reactivity to human microvascular endothelial cells, and used a combination of transcriptomic and proteomic approaches to identify non-HLA Abs. RESULTS: We identified a highly selected cohort of 38 patients with early acute AMVR. Biopsy specimens revealed intense microvascular inflammation and the presence of vasculitis (in 60.5%), interstitial hemorrhages (31.6%), or thrombotic microangiopathy (15.8%). Serum samples collected at the time of transplant showed that previously proposed anti-endothelial cell Abs-angiotensin type 1 receptor (AT1R), endothelin-1 type A and natural polyreactive Abs-did not increase significantly among patients with AMVR compared with a control group of stable kidney transplant recipients. However, 26% of the tested AMVR samples were positive for AT1R Abs when a threshold of 10 IU/ml was used. The crossmatch assay identified a common IgG response that was specifically directed against constitutively expressed antigens of microvascular glomerular cells in patients with AMVR. Transcriptomic and proteomic analyses identified new targets of non-HLA Abs, with little redundancy among individuals. CONCLUSIONS: Our findings indicate that preformed IgG Abs targeting non-HLA antigens expressed on glomerular endothelial cells are associated with early AMVR, and that in vitro cell-based assays are needed to improve risk assessments before transplant.


Asunto(s)
Rechazo de Injerto/inmunología , Hemorragia/inmunología , Inmunoglobulina G/sangre , Receptor de Angiotensina Tipo 1/inmunología , Microangiopatías Trombóticas/inmunología , Vasculitis/inmunología , Enfermedad Aguda , Adulto , Anciano , Células Endoteliales/inmunología , Endotelina-1/inmunología , Femenino , Rechazo de Injerto/patología , Rechazo de Injerto/fisiopatología , Hemorragia/patología , Humanos , Glomérulos Renales/patología , Trasplante de Riñón/efectos adversos , Masculino , Microvasos/patología , Persona de Mediana Edad , Microangiopatías Trombóticas/patología , Factores de Tiempo , Vasculitis/patología
7.
Haematologica ; 103(5): 778-786, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29472357

RESUMEN

Sickle cell disease is characterized by chronic anemia and vaso-occlusive crises, which eventually lead to multi-organ damage and premature death. Hematopoietic stem cell transplantation is the only curative treatment but it is limited by toxicity and poor availability of HLA-compatible donors. A gene therapy approach based on the autologous transplantation of lentiviral-corrected hematopoietic stem and progenitor cells was shown to be efficacious in one patient. However, alterations of the bone marrow environment and properties of the red blood cells hamper the harvesting and immunoselection of patients' stem cells from bone marrow. The use of Filgrastim to mobilize large numbers of hematopoietic stem and progenitor cells into the circulation has been associated with severe adverse events in sickle cell patients. Thus, broader application of the gene therapy approach requires the development of alternative mobilization methods. We set up a phase I/II clinical trial whose primary objective was to assess the safety of a single injection of Plerixafor in sickle cell patients undergoing red blood cell exchange to decrease the hemoglobin S level to below 30%. The secondary objective was to measure the efficiency of mobilization and isolation of hematopoietic stem and progenitor cells. No adverse events were observed. Large numbers of CD34+ cells were mobilized extremely quickly. Importantly, the mobilized cells contained high numbers of hematopoietic stem cells, expressed high levels of stemness genes, and engrafted very efficiently in immunodeficient mice. Thus, Plerixafor can be safely used to mobilize hematopoietic stem cells in sickle cell patients; this finding opens up new avenues for treatment approaches based on gene addition and genome editing. Clinicaltrials.gov identifier: NCT02212535.


Asunto(s)
Anemia de Células Falciformes/terapia , Transfusión Sanguínea , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/administración & dosificación , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/patología , Fármacos Anti-VIH/administración & dosificación , Antígenos CD34/metabolismo , Antidrepanocíticos/administración & dosificación , Bencilaminas , Estudios de Casos y Controles , Células Cultivadas , Estudios de Cohortes , Ciclamas , Células Madre Hematopoyéticas/citología , Humanos , Hidroxiurea/administración & dosificación
8.
Am J Hematol ; 93(6): 794-802, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29603363

RESUMEN

Sickle cell disease (SCD) in pregnancy can be associated with adverse maternal and perinatal outcomes. Furthermore, complications of SCD can be aggravated by pregnancy. Optimal prenatal care aims to decrease the occurrence of maternal and fetal complications. A retrospective, French, two-center study compared two care strategies for pregnant women with SCD over two time periods. In the first study period (2005-2010), the women were systematically offered prophylactic transfusions. In the second study period (2011-2014), a targeted transfusion strategy was applied whenever possible, and home-based prophylactic nocturnal oxygen therapy was offered to all the pregnant women. The two periods did not differ significantly in terms of the incidence of vaso-occlusive events. Maternal mortality, perinatal mortality, and obstetric complication rates were also similar in the two periods, as was the incidence of post-transfusion complications (6.1% in 2005-2010 and 1.3% in 2011-2014, P = .15), although no de novo alloimmunizations or delayed hemolysis transfusion reactions were observed in the second period. The results of this preliminary, retrospective study indicate that targeted transfusion plus home-based prophylactic nocturnal oxygen therapy is safe and may decrease transfusion requirements and transfusion-associated complications.


Asunto(s)
Transfusión Sanguínea/métodos , Oxígeno/uso terapéutico , Reacción a la Transfusión/prevención & control , Adolescente , Adulto , Arteriopatías Oclusivas/etiología , Arteriopatías Oclusivas/prevención & control , Femenino , Francia , Humanos , Embarazo , Complicaciones Hematológicas del Embarazo , Premedicación , Estudios Retrospectivos , Encuestas y Cuestionarios , Adulto Joven
9.
Br J Haematol ; 179(2): 323-335, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28699644

RESUMEN

In African-American patients with sickle cell disease (SCD), APOL1 G1 and G2 variants are associated with increased risk of sickle cell nephropathy (SCN). To determine the role of APOL1 variants in SCD patients living in Europe, we genotyped 152 SCD patients [aged 30·4 (24·3-36·4) years], mainly of Sub-Saharan African ancestry, for APOL1 G1 and G2 and for variants of four genes with kidney tropism (GSTM1, GSTT1, GSTP1, and HMOX1). Homozygous or double-heterozygous APOL G1 and G2 genotypes were strongly associated with end stage renal disease (P = 0·003) and worse Kidney Disease: Improving Global Outcomes stages (P = 0·001). Further, these genotypes were associated in an age-dependent manner with lower estimated glomerular filtration rate (eGFR, P = 0·008), proteinuria (P = 0·009) and albuminuria (P < 0·001) but not with other SCD complications. Compared to APOL1 G1/wild type (WT), the APOL1 G2/WT genotype was associated with a lower eGFR (P = 0·04) in an age-dependent manner, suggesting that the G2/WT patients are likely to have worse kidney prognosis. Other genes variants analysed were not associated with SCN or other SCD complications. Our data indicate that APOL1 screening should be considered for the management of SCD patients, including those of non-African-American origin, as those with homozygous or double heterozygous variants are clearly at higher risk of SCN.


Asunto(s)
Albuminuria , Anemia de Células Falciformes , Apolipoproteínas/genética , Variación Genética , Heterocigoto , Homocigoto , Riñón/fisiopatología , Lipoproteínas HDL/genética , Adulto , Negro o Afroamericano , Albuminuria/genética , Albuminuria/fisiopatología , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/fisiopatología , Apolipoproteína L1 , Femenino , Tasa de Filtración Glomerular , Gutatión-S-Transferasa pi/genética , Glutatión Transferasa/genética , Hemo-Oxigenasa 1/genética , Humanos , Masculino
11.
Am J Hematol ; 92(2): 136-140, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27813144

RESUMEN

Erythrocytapheresis (ER) can improve outcome in patients with sickle cell disease (SCD). A good vascular access is required but frequently it can be difficult to obtain for sickle cell patients. Arterio-venous fistulas (AVFs) have been suggested for ER in SCD supported by limited evidence. We report the largest cohort of ER performed with AVFs from three French SCD reference centers. Data of SCD patients undergoing ER with AVFs in the French SCD reference center were retrospectively collected. The inclusion criteria were: SS or Sß-Thalassemia and AVF surgery for ER. SCD-related complications, transfusion history, details about AVF surgical procedure, echocardiographic data before and after AVF, AVF-related surgical and hemodynamical complications were collected. Twenty-six patients (mean age 20.5 years, mean follow-up 68 months [11-279]) were included. Twenty-three patients (88.5%) required central vascular access before AVF. Fifteen AVFs (58%) were created on the forearm and 11 (42%) on the arm. Nineteen patients (73%) had stenotic, thrombotic or infectious AVF complications. A total of 0.36 stenosis per 1,000 AVF days, 0.37 thrombosis per 1,000 AVF days and 0.078 infections per 1.000 AVF days were observed. The mean AVF lifespan was 51 months [13-218]. One patient with severe pulmonary hypertension worsened after AVF creation and died. We report the first series of SCD patients with AVF for ER, demonstrating that AVFs could be considered as a potential vascular access for ER. Patients with increased risk for hemodynamic intolerance of AVFs must be carefully identified, so that alternative vascular accesses can be considered. Am. J. Hematol. 92:136-140, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anemia de Células Falciformes/terapia , Derivación Arteriovenosa Quirúrgica/métodos , Eliminación de Componentes Sanguíneos/métodos , Transfusión de Eritrocitos/métodos , Adolescente , Adulto , Anemia de Células Falciformes/sangre , Derivación Arteriovenosa Quirúrgica/efectos adversos , Eliminación de Componentes Sanguíneos/efectos adversos , Estudios de Cohortes , Constricción Patológica/epidemiología , Constricción Patológica/etiología , Transfusión de Eritrocitos/efectos adversos , Femenino , Humanos , Hierro/sangre , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Trombosis/epidemiología , Trombosis/etiología , Resultado del Tratamiento , Adulto Joven
12.
J Am Soc Nephrol ; 27(8): 2520-7, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26701979

RESUMEN

FSGS is a common glomerular disorder that has a high propensity for recurrence after kidney transplant. The pathophysiology of FSGS is unknown, but podocytes seem to be the target of one or several circulating factors that lead to cytoskeleton reorganization and proteinuria. Research on podocytes has identified B7-1 as an important factor in podocyte biology and a new therapeutic target in renal disease. Indeed, in four patients with recurrent FSGS after transplant, treatment with the B7-1 blocker abatacept was associated with proteinuria remission. Here, we prospectively treated nine patients with recurrent FSGS after transplant using either abatacept or belatacept, a B7-1 blocker with higher affinity, and did not induce proteinuria remission. Furthermore, we did not detect B7-1 expression by immunofluorescence in podocytes of biopsy specimens from these or other kidney grafts or podocytes of native kidney biopsy specimens. In conclusion, B7-1 blockade did not induce FSGS remission after transplant in our study.


Asunto(s)
Abatacept/farmacología , Abatacept/uso terapéutico , Antígeno B7-1/antagonistas & inhibidores , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Trasplante de Riñón , Síndrome Nefrótico/etiología , Síndrome Nefrótico/prevención & control , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/prevención & control , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Recurrencia , Insuficiencia del Tratamiento , Adulto Joven
13.
Kidney Int ; 90(5): 1037-1044, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27528551

RESUMEN

Recent research on podocytes has proposed B7-1 as an important player in podocyte biology and as a potential new therapeutic target. B7-1 was upregulated in injured podocytes and described as a biomarker to identify patients who may benefit from abatacept, a B7-1 blocker. However, after this initial enthusiasm, several reports have not confirmed the efficiency of abatacept at inducing proteinuria remission in patients. In order to resolve these discrepancies, we explored the role of B7-1 in the injured podocyte. Both primary cultured and immortalized podocytes were exposed to lipopolysaccharides, but this failed to induce B7-1 expression at the mRNA and protein levels. Importantly, TLR-4 engagement confirmed lipopolysaccharide efficacy. We then evaluated B7-1 expression in several mouse models of podocyte injury including treatment with lipopolysaccharide or Adriamycin, a lupus prone model (NZB/W F1) and subtotal nephrectomy. Using 3 commercially available anti-B7-1 antibodies and appropriate controls, we could not find B7-1 expression in podocytes, whereas some infiltrating cells were positive. Thus, our findings do not support a role for B7-1 in podocyte biology. Hence, further studies are mandatory before treating proteinuric patients with B7-1 blockers.


Asunto(s)
Antígeno B7-1/metabolismo , Podocitos/metabolismo , Animales , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Cultivo Primario de Células
14.
J Am Soc Nephrol ; 26(5): 1161-71, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25381426

RESUMEN

The initial contact point between a recipient's immune system and a transplanted graft is the vascular endothelium. Clinical studies suggest a pathogenic role for non-HLA antiendothelial cell antibodies (AECAs) in allograft rejection; however, evidence linking AECAs of known specificity to in vivo vascular injury is lacking. Here, we used high-density protein arrays to identify target antigens for AECAs isolated from the sera of recipients of kidney transplants experiencing antibody-mediated rejection in the absence of donor-specific HLA antibodies. Four antigenic targets expressed on endothelial cells were identified: endoglin, Fms-like tyrosine kinase-3 ligand, EGF-like repeats and discoidin I-like domains 3, and intercellular adhesion molecule 4; the first three have been implicated in endothelial cell activation and leukocyte extravasation. To validate these findings, ELISAs were constructed, and sera from an additional 150 renal recipients were tested. All four AECAs were detected in 24% of pretransplant sera, and they were associated with post-transplant donor-specific HLA antibodies, antibody-mediated rejection, and early transplant glomerulopathy. AECA stimulation of endothelial cell cultures increased adhesion molecule expression and production of inflammatory cytokines: regulated on activation, normal T cell expressed and secreted PDGF and RESISTIN. These correlations between in vitro experiments and in vivo histopathology suggest that AECAs activate the vascular endothelium, amplifying the alloimmune response and increasing microvascular damage. Given the growing number of transplant candidates, a better understanding of the antigenic targets, beyond HLA, and mechanisms of immune injury will be essential for improving long-term allograft survival.


Asunto(s)
Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Rechazo de Injerto/inmunología , Trasplante de Riñón , Adulto , Anciano , Anticuerpos/sangre , Antígenos/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Rechazo de Injerto/sangre , Antígenos HLA/inmunología , Humanos , Masculino , Persona de Mediana Edad , Análisis por Matrices de Proteínas , Proteómica/métodos , Estudios Retrospectivos
15.
J Am Soc Nephrol ; 26(11): 2840-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25948873

RESUMEN

Urinary levels of C-X-C motif chemokine 9 (CXCL9) and CXCL10 can noninvasively diagnose T cell-mediated rejection (TCMR) of renal allografts. However, performance of these molecules as diagnostic/prognostic markers of antibody-mediated rejection (ABMR) is unknown. We investigated urinary CXCL9 and CXCL10 levels in a highly sensitized cohort of 244 renal allograft recipients (67 with preformed donor-specific antibodies [DSAs]) with 281 indication biopsy samples. We assessed the benefit of adding these biomarkers to conventional models for diagnosing/prognosing ABMR. Urinary CXCL9 and CXCL10 levels, normalized to urine creatinine (Cr) levels (CXCL9:Cr and CXCL10:Cr) or not, correlated with the extent of tubulointerstitial (i+t score; all P<0.001) and microvascular (g+ptc score; all P<0.001) inflammation. CXCL10:Cr diagnosed TCMR (area under the curve [AUC]=0.80; 95% confidence interval [95% CI], 0.68 to 0.92; P<0.001) and ABMR (AUC=0.76; 95% CI, 0.69 to 0.82; P<0.001) with high accuracy, even in the absence of tubulointerstitial inflammation (AUC=0.70; 95% CI, 0.61 to 0.79; P<0.001). Although mean fluorescence intensity of the immunodominant DSA diagnosed ABMR (AUC=0.75; 95% CI, 0.68 to 0.82; P<0.001), combining urinary CXCL10:Cr with immunodominant DSA levels improved the diagnosis of ABMR (AUC=0.83; 95% CI, 0.77 to 0.89; P<0.001). At the time of ABMR, urinary CXCL10:Cr ratio was independently associated with an increased risk of graft loss. In conclusion, urinary CXCL10:Cr ratio associates with tubulointerstitial and microvascular inflammation of the renal allograft. Combining the urinary CXCL10:Cr ratio with DSA monitoring significantly improves the noninvasive diagnosis of ABMR and the stratification of patients at high risk for graft loss.


Asunto(s)
Quimiocina CXCL10/orina , Rechazo de Injerto , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/inmunología , Adulto , Anticuerpos/sangre , Área Bajo la Curva , Biomarcadores/orina , Biopsia , Quimiocina CXCL9/orina , Estudios de Cohortes , Creatinina/orina , Femenino , Humanos , Inflamación , Interferón gamma/metabolismo , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Insuficiencia Renal/orina , Reproducibilidad de los Resultados , Trasplante Homólogo
17.
Nat Commun ; 12(1): 6446, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750385

RESUMEN

The use of chimeric antigen receptor (CAR)-engineered regulatory T cells (Tregs) has emerged as a promising strategy to promote immune tolerance. However, in conventional T cells (Tconvs), CAR expression is often associated with tonic signaling, which can induce CAR-T cell dysfunction. The extent and effects of CAR tonic signaling vary greatly according to the expression intensity and intrinsic properties of the CAR. Here, we show that the 4-1BB CSD-associated tonic signal yields a more dramatic effect in CAR-Tregs than in CAR-Tconvs with respect to activation and proliferation. Compared to CD28 CAR-Tregs, 4-1BB CAR-Tregs exhibit decreased lineage stability and reduced in vivo suppressive capacities. Transient exposure of 4-1BB CAR-Tregs to a Treg stabilizing cocktail, including an mTOR inhibitor and vitamin C, during ex vivo expansion sharply improves their in vivo function and expansion after adoptive transfer. This study demonstrates that the negative effects of 4-1BB tonic signaling in Tregs can be mitigated by transient mTOR inhibition.


Asunto(s)
Receptores Quiméricos de Antígenos/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Inmunosupresores/farmacología , Inmunoterapia Adoptiva/métodos , Células Jurkat , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Trasplante Heterólogo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
18.
Nephrol Ther ; 15 Suppl 1: S53-S59, 2019 04.
Artículo en Francés | MEDLINE | ID: mdl-30981396

RESUMEN

In solid organ transplantation, the deleterious effect of antibodies directed against donor HLA antigens, whether preformed or de novo, is well established. Anti-HLA antibodies have been associated not only with the risk of antibody-mediated rejection but also with late graft dysfunction and are now considered to be the leading cause of allograft loss after renal transplantation. In addition to HLA antibodies, the possible involvement of non-HLA antibodies targeting donor endothelial cells has long been the subject of intense research. The purpose of this review is to discuss current knowledge and remaining issues related to the involvement of non-HLA antibodies in solid organ transplantation. More specifically, the clinical data underlying the hypothesis of the role of non-HLA antibodies will be discussed, as well as the different techniques for antibody detection, their clinical relevance and their antigenic targets.

19.
Hum Immunol ; 80(8): 614-620, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31054781

RESUMEN

Anti-endothelial cell antibodies (AECAs) have been correlated with increased acute and chronic rejection across all organ types and early graft dysfunction in kidney and heart transplantation. Nevertheless, the lack of appropriate tools and clear criteria for defining injurious versus non-injurious AECAs prohibits their routine inclusion in clinical risk assessments and diagnostic algorithms for antibody mediated injury. Clinical characterization of AECAs is complicated due to the wide range of polymorphic and non-polymorphic antigens expressed across different vascular tissues and the diverse array of specificities observed between individuals. This complexity is also reflected in the broad spectrum of reported injury phenotypes. AECAs detected at time of allograft dysfunction may represent biomarkers of past vascular injury or active contributors to a current rejection process. New tools within the fields of proteomics, genomics, bioinformatics, and imaging are currently being validated and hold great promise for unraveling the AECA paradox.


Asunto(s)
Anticuerpos/metabolismo , Biomarcadores/metabolismo , Células Endoteliales/inmunología , Rechazo de Injerto/inmunología , Trasplante de Órganos , Animales , Autoantígenos/inmunología , Exosomas/metabolismo , Humanos , Inmunización , Isoantígenos/inmunología , Especificidad de Órganos
20.
Nephrol Ther ; 15(6): 469-484, 2019 Nov.
Artículo en Francés | MEDLINE | ID: mdl-31640943

RESUMEN

More than fifty years after the success of the two first renal transplantations in Boston and in Necker hospital in Paris, renal transplantation became the treatment of choice of end stage renal failure, because it improves not only the quality of life of the patients but also their long-term survival. In France, more than 3,700 kidney transplantations are performed every year and more than 40,000 patients are living with a functioning kidney allograft. This treatment of end stage renal disease requires a fine-tuned pre-transplant evaluation and a multidisciplinary post-transplant care in order to prevent, to detect and to treat comorbidities and complications of immunosuppression. The ambition of this manuscript is not to describe in an exhaustive way all the aspects of renal transplantation but starting from the experience of a team, recently published data, and national and international guidelines, to try to provide a synthetic and chronological view of the early post-transplant monitoring.


Asunto(s)
Trasplante de Riñón , Cuidados Posteriores , Biopsia/métodos , Contraindicaciones de los Procedimientos , Funcionamiento Retardado del Injerto , Estudios de Seguimiento , Tasa de Filtración Glomerular , Rechazo de Injerto/prevención & control , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/uso terapéutico , Consentimiento Informado , Riñón/patología , Fallo Renal Crónico/cirugía , Trasplante de Riñón/métodos , Complicaciones Posoperatorias , Guías de Práctica Clínica como Asunto , Cuidados Preoperatorios , Donantes de Tejidos , Obtención de Tejidos y Órganos , Trasplantes/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA